Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
RNA ; 26(11): 1731-1742, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32759389

RESUMEN

The measurement of RNA abundance derived from massively parallel sequencing experiments is an essential technique. Methods that reduce ribosomal RNA levels are usually required prior to sequencing library construction because ribosomal RNA typically comprises the vast majority of a total RNA sample. For some experiments, ribosomal RNA depletion is favored over poly(A) selection because it offers a more inclusive representation of the transcriptome. However, methods to deplete ribosomal RNA are generally proprietary, complex, inefficient, applicable to only specific species, or compatible with only a narrow range of RNA input levels. Here, we describe Ribo-Pop (ribosomal RNA depletion for popular use), a simple workflow and antisense oligo design strategy that we demonstrate works over a wide input range and can be easily adapted to any organism with a sequenced genome. We provide a computational pipeline for probe selection, a streamlined 20-min protocol, and ready-to-use oligo sequences for several organisms. We anticipate that our simple and generalizable "open source" design strategy would enable virtually any laboratory to pursue full transcriptome sequencing in their organism of interest with minimal time and resource investment.


Asunto(s)
Biología Computacional/métodos , Oligorribonucleótidos Antisentido/genética , ARN Ribosómico/análisis , Secuencia de Bases , Biología Computacional/economía , Análisis Costo-Beneficio , Secuenciación de Nucleótidos de Alto Rendimiento , Sondas de Oligonucleótidos/genética , ARN Ribosómico/antagonistas & inhibidores , Análisis de Secuencia de ARN/métodos , Flujo de Trabajo
2.
Biochemistry ; 56(38): 5049-5052, 2017 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-28782938

RESUMEN

We report the discovery of the first bacterial ribosomal RNA (rRNA) synthesis inhibitor that has specific antimicrobial activity against methicillin-resistant Staphylococcus aureus (MRSA). A pharmacophore model was constructed on the basis of the protein-protein interaction between essential bacterial rRNA transcription factors NusB and NusE and employed for an in silico screen to identify potential leads. One compound, (E)-2-{[(3-ethynylphenyl)imino]methyl}-4-nitrophenol (MC4), demonstrated antimicrobial activity against a panel of S. aureus strains, including MRSA, without significant toxicity to mammalian cells. MC4 resulted in a decrease in the rRNA level in bacteria, and the target specificity of MC4 was confirmed at the molecular level. Results obtained from this work validated the bacterial rRNA transcription machinery as a novel antimicrobial target. This approach may be extended to other factors in rRNA transcription, and MC4 could be applied as a chemical probe to dissect the relationship among MRSA infection, MRSA growth rate, and rRNA synthesis, in addition to its therapeutic potential.


Asunto(s)
Antibacterianos/farmacología , Hidrazonas/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Nitrofenoles/farmacología , ARN Ribosómico/antagonistas & inhibidores , Antibacterianos/efectos adversos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Simulación por Computador , Evaluación Preclínica de Medicamentos/métodos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Hidrazonas/química , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Pruebas de Sensibilidad Microbiana , Nitrofenoles/química , Conformación Proteica , ARN Ribosómico/biosíntesis , ARN Ribosómico/genética , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo
3.
Biopolymers ; 107(4)2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27858985

RESUMEN

The bacterial ribosome has many functional ribosomal RNA (rRNA) sites. We have computationally analyzed the rRNA regions involved in the interactions between the 30S and 50S subunits. Various properties of rRNA such as solvent accessibility, opening energy, hydrogen bonding pattern, van der Waals energy, thermodynamic stability were determined. Based on these properties we selected rRNA targets for hybridization with complementary 2'-O-methyl oligoribonucleotides (2'-OMe RNAs). Further, the inhibition efficiencies of the designed ribosome-interfering 2'-OMe RNAs were tested using a ß-galactosidase assay in a translation system based on the E. coli extract. Several of the oligonucleotides displayed IC50 values below 1 µM, which were in a similar range as those determined for known ribosome inhibitors, tetracycline and pactamycin. The calculated opening and van der Waals stacking energies of the rRNA targets correlated best with the inhibitory efficiencies of 2'-OMe RNAs. Moreover, the binding affinities of several oligonucleotides to both 70S ribosomes and isolated 30S and 50S subunits were measured using a double-filter retention assay. Further, we applied heat-shock chemical transformation to introduce 2'-OMe RNAs to E. coli cells and verify inhibition of bacterial growth. We observed high correlation between IC50 in the cell-free extract and bacterial growth inhibition. Overall, the results suggest that the computational analysis of potential rRNA targets within the conformationally dynamic regions of inter-subunit bridges can help design efficient antisense oligomers to probe the ribosome function.


Asunto(s)
Oligonucleótidos/metabolismo , ARN Ribosómico/metabolismo , Secuencia de Bases , Sitios de Unión , Diseño Asistido por Computadora , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Conformación de Ácido Nucleico , Oligonucleótidos/química , Pactamicina/química , Pactamicina/metabolismo , Pactamicina/farmacología , Unión Proteica , Biosíntesis de Proteínas/efectos de los fármacos , Estructura Terciaria de Proteína , ARN Ribosómico/antagonistas & inhibidores , ARN Ribosómico/química , Subunidades Ribosómicas Grandes Bacterianas/química , Subunidades Ribosómicas Grandes Bacterianas/metabolismo , Subunidades Ribosómicas Pequeñas Bacterianas/química , Subunidades Ribosómicas Pequeñas Bacterianas/metabolismo
4.
Int J Mol Sci ; 18(1)2017 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-28117679

RESUMEN

Overall survival for patients with ovarian cancer (OC) has shown little improvement for decades meaning new therapeutic options are critical. OC comprises multiple histological subtypes, of which the most common and aggressive subtype is high-grade serous ovarian cancer (HGSOC). HGSOC is characterized by genomic structural variations with relatively few recurrent somatic mutations or dominantly acting oncogenes that can be targeted for the development of novel therapies. However, deregulation of pathways controlling homologous recombination (HR) and ribosome biogenesis has been observed in a high proportion of HGSOC, raising the possibility that targeting these basic cellular processes may provide improved patient outcomes. The poly (ADP-ribose) polymerase (PARP) inhibitor olaparib has been approved to treat women with defects in HR due to germline BRCA mutations. Recent evidence demonstrated the efficacy of targeting ribosome biogenesis with the specific inhibitor of ribosomal RNA synthesis, CX-5461 in v-myc avian myelocytomatosis viral oncogene homolog (MYC)-driven haematological and prostate cancers. CX-5461 has now progressed to a phase I clinical trial in patients with haematological malignancies and phase I/II trial in breast cancer. Here we review the currently available targeted therapies for HGSOC and discuss the potential of targeting ribosome biogenesis as a novel therapeutic approach against HGSOC.


Asunto(s)
Benzotiazoles/uso terapéutico , Cistadenocarcinoma Seroso/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Naftiridinas/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , ARN Ribosómico/antagonistas & inhibidores , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Femenino , Humanos , Modelos Genéticos , Terapia Molecular Dirigida/tendencias , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Ftalazinas/uso terapéutico , Piperazinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , ARN Ribosómico/genética , ARN Ribosómico/metabolismo
5.
Blood ; 124(4): 579-89, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-24850755

RESUMEN

The transcription initiation factor I (TIF-IA) is an important regulator of the synthesis of ribosomal RNA (rRNA) through its facilitation of the recruitment of RNA polymerase I (Pol I) to the ribosomal DNA promoter. Activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, which occurs commonly in acute myelogenous leukemia, enhances rRNA synthesis through TIF-IA stabilization and phosphorylation. We have discovered that TIF-IA coexists with a splicing isoform, TIF-90, which is expressed preferentially in the nucleolus and at higher levels in proliferating and transformed hematopoietic cells. TIF-90 interacts directly with Pol I to increase rRNA synthesis as a consequence of Akt activation. Furthermore, TIF-90 binds preferentially to a 90-kDa cleavage product of the actin binding protein filamin A (FLNA) that inhibits rRNA synthesis. Increased expression of TIF-90 overcomes the inhibitory effect of this cleavage product and stimulates rRNA synthesis. Because activated Akt also reduces FLNA cleavage, these results indicate that activated Akt and TIF-90 function in parallel to increase rRNA synthesis and, as a consequence, cell proliferation in leukemic cells. These results provide evidence that the direct targeting of Akt would be an effective therapy in acute leukemias in which Akt is activated.


Asunto(s)
Empalme Alternativo , Proteínas Reguladoras de la Apoptosis/metabolismo , Filaminas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Nucleares/metabolismo , ARN Ribosómico/metabolismo , Ribosomas/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Western Blotting , Núcleo Celular/genética , Proliferación Celular , Inmunoprecipitación de Cromatina , Filaminas/genética , Humanos , Leucemia Mieloide Aguda/genética , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Ribosómico/antagonistas & inhibidores , ARN Ribosómico/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Células Tumorales Cultivadas
6.
J Cell Sci ; 124(Pt 17): 3017-28, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21878508

RESUMEN

The tumour suppressor p53 negatively controls cell cycle progression in response to perturbed ribosome biogenesis in mammalian cells, thus coordinating growth with proliferation. Unlike mammalian cells, p53 is not involved in the growth control of proliferation in yeasts and flies. We investigated whether a p53-independent mechanism of response to inadequate ribosome biogenesis rate is also present in mammalian cells. We studied the effect of specific inhibition of rRNA synthesis on cell cycle progression in human cancer cell lines using the small-interfering RNA procedure to silence the POLR1A gene, which encodes the catalytic subunit of RNA polymerase I. We found that interference of POLR1A inhibited the synthesis of rRNA and hindered cell cycle progression in cells with inactivated p53, as a consequence of downregulation of the transcription factor E2F-1. Downregulation of E2F-1 was due to release of the ribosomal protein L11, which inactivated the E2F-1-stabilising function of the E3 ubiquitin protein ligase MDM2. These results demonstrated the existence of a p53-independent mechanism that links cell growth to cell proliferation in mammalian cells, and suggested that selective targeting of the RNA polymerase I transcription machinery might be advisable to hinder proliferation of p53-deficient cancer cells.


Asunto(s)
Factor de Transcripción E2F1/genética , ARN Polimerasa I/genética , ARN Ribosómico/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Dominio Catalítico , Ciclo Celular/genética , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo , Factor de Transcripción E2F1/metabolismo , Silenciador del Gen , Células HCT116 , Humanos , ARN Ribosómico/biosíntesis , ARN Ribosómico/genética , Transcripción Genética , Transfección , Proteína p53 Supresora de Tumor/metabolismo
7.
Int J Mol Sci ; 14(8): 15532-45, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23892598

RESUMEN

Ribosome-inactivating proteins (RIPs), enzymes that are widely distributed in the plant kingdom, inhibit protein synthesis by depurinating rRNA and many other polynucleotidic substrates. Although RIPs show antiviral, antifungal, and insecticidal activities, their biological and physiological roles are not completely understood. Additionally, it has been described that RIP expression is augmented under stressful conditions. In this study, we evaluated protein synthesis inhibition activity in partially purified basic proteins (hereafter referred to as RIP activity) from tissue extracts of Fragaria × ananassa (strawberry) cultivars with low (Dora) and high (Record) tolerance to root pathogens and fructification stress. Association between the presence of RIP activity and the crop management (organic or integrated soil), growth stage (quiescence, flowering, and fructification), and exogenous stress (drought) were investigated. RIP activity was found in every tissue tested (roots, rhizomes, leaves, buds, flowers, and fruits) and under each tested condition. However, significant differences in RIP distribution were observed depending on the soil and growth stage, and an increase in RIP activity was found in the leaves of drought-stressed plants. These results suggest that RIP expression and activity could represent a response mechanism against biotic and abiotic stresses and could be a useful tool in selecting stress-resistant strawberry genotypes.


Asunto(s)
Fragaria/enzimología , Fragaria/metabolismo , Extractos Vegetales/metabolismo , Proteínas Inactivadoras de Ribosomas/metabolismo , Animales , Estadios del Ciclo de Vida , Biosíntesis de Proteínas , ARN Ribosómico/antagonistas & inhibidores , ARN Ribosómico/metabolismo , Estrés Fisiológico
8.
J Biol Chem ; 285(47): 36857-64, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20843798

RESUMEN

Solid tumor development is frequently accompanied by energy-deficient conditions such as glucose deprivation and hypoxia. Follistatin (FST), a secretory protein originally identified from ovarian follicular fluid, has been suggested to be involved in tumor development. However, whether it plays a role in cancer cell survival under energy-deprived conditions remains elusive. In this study, we demonstrated that glucose deprivation markedly enhanced the expression and nucleolar localization of FST in HeLa cells. The nucleolar localization of FST relied on its nuclear localization signal (NLS) comprising the residues 64-87. Localization of FST to the nucleolus attenuated rRNA synthesis, a key process for cellular energy homeostasis and cell survival. Overexpression of FST delayed glucose deprivation-induced apoptosis, whereas down-regulation of FST exerted the opposite effect. These functions depended on the presence of an intact NLS because the NLS-deleted mutant of FST lost the rRNA inhibition effect and the cell protective effect. Altogether, we identified a novel nucleolar function of FST, which is of importance in the modulation of cancer cell survival in response to glucose deprivation.


Asunto(s)
Apoptosis , Nucléolo Celular/metabolismo , Folistatina/metabolismo , Glucosa/deficiencia , ARN Ribosómico/biosíntesis , Neoplasias del Cuello Uterino/patología , Northern Blotting , Western Blotting , Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Femenino , Técnica del Anticuerpo Fluorescente , Folistatina/antagonistas & inhibidores , Folistatina/genética , Células HeLa , Humanos , Señales de Localización Nuclear , ARN Mensajero/genética , ARN Ribosómico/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias del Cuello Uterino/metabolismo
9.
J Biol Chem ; 285(47): 36260-6, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20720019

RESUMEN

Calcineurin homologous protein 1 (CHP1) is a widely expressed, 22-kDa myristoylated EF-hand Ca(2+)-binding protein that shares a high degree of similarity with the regulatory B subunit of calcineurin (65%) and with calmodulin (59%). CHP1 localizes to the plasma membrane, the Golgi apparatus, and the nucleus and functions to regulate trafficking of early secretory vesicles, activation of T cells, and expression and transport of the Na-H exchanger NHE1. Although CHP1 contains nuclear export signals, whether its nuclear and cytoplasmic localization is regulated and has distinct functions remain unknown. We show that CHP1 is predominantly in the nucleus in quiescent fibroblasts, is translocated to cytoplasmic compartments with growth medium, and that translocation is inhibited by mutations in the nuclear export motifs. In a screen for proteins co-precipitating with CHP1 in quiescent cells we identified the upstream binding factor UBF, a DNA-binding protein and component of the RNA polymerase I complex regulating RNA synthesis. The CHP1-UBF interaction is restricted to the nucleus and inhibited by Ca(2+). Nuclear retention of CHP1 attenuates the abundance of UBF in the nucleolus and inhibits RNA synthesis when quiescent cells are transferred to growth medium. These data show UBF as a newly identified CHP1-binding protein and regulation of RNA synthesis as a newly identified function for nuclear-localized CHP1, which is distinct from CHP1 functions in the cytosol.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Núcleo Celular/metabolismo , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , ARN Ribosómico/antagonistas & inhibidores , Ribosomas/metabolismo , Animales , Sitios de Unión , Western Blotting , Proteínas de Unión al Calcio/genética , Membrana Celular/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , Cricetinae , Citoplasma/metabolismo , Fibroblastos/metabolismo , Aparato de Golgi/metabolismo , Pulmón/citología , Pulmón/metabolismo , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Unión Proteica , Transporte de Proteínas , ARN Mensajero/genética , ARN Ribosómico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
10.
Cell Mol Life Sci ; 67(3): 335-40, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19921484

RESUMEN

The expression of the murine p200 family protein p204 in numerous tissues can be activated by a variety of distinct, tissue-specific transcription factors. p204 modulates cell proliferation, cell cycling, and the differentiation of various tissues, including skeletal muscle myotubes, beating cardiac myocytes, osteoblasts, chondrocytes, and macrophages. This protein modulates these processes in various ways, such as by (1) blocking ribosomal RNA synthesis in the nucleolus, (2) inhibiting Ras signaling in the cytoplasm, (3) promoting the activity of particular transcription factors in the nucleus by forming complexes with them, and (4) overcoming the block of the activity of other transcription factors by inhibitor of differentiation (Id) proteins. Much remains to be learned about p204, particularly with respect to its expected involvement in the differentiation of several as yet unexplored tissues.


Asunto(s)
Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Condrocitos/citología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Mioblastos Esqueléticos/citología , Miocitos Cardíacos/citología , Proteínas Nucleares/fisiología , Osteoblastos/citología , Fosfoproteínas/fisiología , ARN Ribosómico/antagonistas & inhibidores , ARN Ribosómico/metabolismo , Proteínas ras/antagonistas & inhibidores , Proteínas ras/metabolismo
11.
J Am Chem Soc ; 132(20): 6973-81, 2010 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-20441189

RESUMEN

Ribosomally produced thiopeptide antibiotics are highly promising lead compounds targeting the GTPase-associated region (GAR) of the bacterial ribosome. A representative panel of GAR mutants suspected to confer resistance against thiopeptide antibiotics was reconstituted in vitro and quantitatively studied with fluorescent probes. It was found that single-site mutations of the ribosomal 23S rRNA binding site region directly affect thiopeptide affinity. Quantitative equilibrium binding data clearly identified A1067 as the base contributing most strongly to the binding environment. The P25 residue on the ribosomal protein L11 was essential for binding of the monocyclic thiopeptides micrococcin and promothiocin B, confirming that the mutation of this residue in the producer organism confers self-resistance. For the bicyclic thiopeptides thiostrepton and nosiheptide, all studied single-site resistance mutations on the L11 protein were still fully capable of ligand binding in the upper pM range, both in the RNA-protein complex and in isolated 70S ribosomes. These single-site mutants were then specifically reconstituted in Bacillus subtilis, confirming their efficacy as resistance-conferring. It is thus reasoned that, in contrast to modifications of the 23S rRNA in the GAR, mutations of the L11 protein do not counteract binding of bicyclic thiopeptides, but allow the ribosome to bypass the protein biosynthesis blockade enforced by these antibiotics in the wild type.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Farmacorresistencia Bacteriana , Péptidos/farmacología , Secuencia de Aminoácidos , Antibacterianos/química , Antibacterianos/metabolismo , Bacterias/citología , Bacterias/genética , Bacterias/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Modelos Moleculares , Mutación , Péptidos/química , Péptidos/metabolismo , Conformación Proteica , ARN Bacteriano/antagonistas & inhibidores , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Ribosómico/antagonistas & inhibidores , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Subunidades Ribosómicas Grandes Bacterianas/efectos de los fármacos , Subunidades Ribosómicas Grandes Bacterianas/genética
12.
J Gen Virol ; 91(Pt 5): 1239-44, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20089798

RESUMEN

Both entero- and cardioviruses have been shown to suppress host mRNA synthesis. Enteroviruses are also known to inhibit the activity of rRNA genes, whereas this ability of cardioviruses is under debate. This study reported that mengovirus (a cardiovirus) suppressed rRNA synthesis but less efficiently than poliovirus (an enterovirus). In contrast to poliovirus infection, the incorporation of BrUTP, fluorouridine and [14C]uridine in rRNA precursors was observed even during the late stages of mengovirus infection, although at a significantly reduced level. The cleavage of TATA-binding protein, considered to be one of the central events in poliovirus-induced transcription shutoff, was not detected in mengovirus-infected cells, indicating a difference in the mechanisms of host RNA synthesis inhibition caused by these viruses. The results also showed that functional leader protein is redundant for the suppression of host RNA synthesis by cardiovirus.


Asunto(s)
Interacciones Huésped-Patógeno , Mengovirus/patogenicidad , Poliovirus/patogenicidad , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , ARN Ribosómico/antagonistas & inhibidores , ARN Ribosómico/biosíntesis , Células HeLa , Humanos
13.
Biochem Biophys Res Commun ; 394(1): 42-7, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20171949

RESUMEN

Currently, [(3)H]uridine is most often used to monitor rRNA synthesis in cultured cells. We show here that radiolabeled ribonucleoside triphosphates, such as [alpha-(33)P]UTP, in culture medium were also incorporated efficiently not only into cells but also into de novo RNA, particularly rRNA. Using this method, we first revealed that endoplasmic reticulum (ER) stress inducers such as tunicamycin and thapsigargin suppressed de novo rRNA synthesis, and that PERK, but not IRE1alpha or ATF6, mediated the suppression. PERK is known to mediate the suppression of de novo protein synthesis via phosphorylation of eIF2alpha. Consistently, other translational inhibitors such as PSI, proteasomal inhibitor, and cycloheximide suppressed de novo rRNA synthesis. eIF2alpha knockdown also suppressed both de novo protein and rRNA syntheses. Furthermore, ER stress reduced cellular ATP levels, and the suppression of rRNA synthesis apparently mitigated their reduction. These observations provided a close link between ATP levels and suppression of de novo rRNA synthesis at ER stress, and we proposed a novel feedback mechanism, in which ATP levels were maintained via suppression of de novo rRNA synthesis in ATP-demanding stresses, such as ER stress.


Asunto(s)
Adenosina Trifosfato/metabolismo , Retículo Endoplásmico/enzimología , ARN Ribosómico/biosíntesis , Estrés Fisiológico , eIF-2 Quinasa/metabolismo , Células HeLa , Humanos , Inhibidores de Proteínas Quinasas/farmacología , ARN Ribosómico/antagonistas & inhibidores , Tapsigargina/farmacología , Tunicamicina/farmacología , eIF-2 Quinasa/antagonistas & inhibidores
14.
J Cell Biol ; 53(3): 611-23, 1972 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-4112854

RESUMEN

This study is concerned with the fate of the nucleolar contents, particularly nucleolar RNA, during mitosis Mitotic cells harvested from monolayer cultures of Chinese hamster embryonal cells, KB6 (human) cells, or L929 (mouse) cells were allowed to proceed into interphase in the presence or absence (control) of 0.04-0 08 microg/ml of actinomycin D, a concentration which preferentially inhibits nucleolar (ribosomal) RNA synthesis 3 hr after mitosis, control cells had large, irregularly shaped nucleoli which stained intensely for RNA with azure B and for protein with fast green. In cells which had returned to interphase in the presence of actinomycin D, nucleoli were segregated into two components easily resolvable in the light microscope, and one of these components stained intensely for RNA with azure B. Both nucleolar components stained for protein with fast green In parallel experiments, cultures were incubated with 0.04-0 08 microg/ml actinomycin D for 3 hr before harvesting of mitotic cells, then mitotic cells were washed and allowed to return to interphase in the absence of actinomycin D. 3 hr after mitosis, nuclei of such cells were devoid of large RNA-containing structures, though small, refractile nucleolus-like bodies were observed by phase-contrast microscopy or in material stained for total protein. These experiments indicate that nucleolar RNA made several hours before mitosis persists in the mitotic cell and repopulates nucleoli when they reform after mitosis


Asunto(s)
Nucléolo Celular/metabolismo , Mitosis , ARN Ribosómico/biosíntesis , Animales , Carcinoma , Línea Celular , Nucléolo Celular/análisis , Nucléolo Celular/efectos de los fármacos , Núcleo Celular/análisis , Cricetinae/embriología , Dactinomicina/farmacología , Desoxirribonucleasas , Histocitoquímica , Humanos , Células L , Ratones , Microscopía Electrónica , Microscopía de Contraste de Fase , Mitosis/efectos de los fármacos , Neoplasias de la Boca , Proteínas/análisis , ARN Ribosómico/análisis , ARN Ribosómico/antagonistas & inhibidores , Ribonucleasas , Coloración y Etiquetado , Factores de Tiempo
15.
J Cell Biol ; 59(3): 669-76, 1973 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-4796877

RESUMEN

The effect of various antimetabolites on nuclear pore formation was studied in synchronized HeLa S(3) cells. The nuclear size was determined by light microscopy and the pore number per unit area of nuclear surface by the freeze-etching technique and electron microscopy. It was found that the inhibition of DNA replication or ribosomal RNA synthesis has no effect on nuclear size increase or pore formation. However, the inhibition of ATP synthesis effectively stops nuclear pore formation. Cycloheximide blocks nuclear pore formation at the same time during G(1) phase of the cell cycle when nuclear size increase is blocked by high concentrations of actinomycin D. This suggests that certain proteins or other factors leading to pore formation and nuclear size increase are transcribed and synthesized at about 3-4 h after mitosis, i.e., about 1-2 h before S phase begins.


Asunto(s)
Antimetabolitos/farmacología , Núcleo Celular/metabolismo , Células HeLa/efectos de los fármacos , Adenosina Trifosfato/antagonistas & inhibidores , Adenosina Trifosfato/biosíntesis , Animales , Radioisótopos de Carbono , Bovinos , Línea Celular , Núcleo Celular/efectos de los fármacos , Cicloheximida/farmacología , ADN/antagonistas & inhibidores , ADN/metabolismo , Replicación del ADN/efectos de los fármacos , Dactinomicina/farmacología , Femenino , Células HeLa/metabolismo , Humanos , Mitosis , Tamaño de la Partícula , Biosíntesis de Proteínas , Proteínas/antagonistas & inhibidores , ARN/antagonistas & inhibidores , ARN/biosíntesis , ARN Ribosómico/antagonistas & inhibidores , Timidina/metabolismo , Factores de Tiempo
16.
J Control Release ; 286: 1-9, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30016731

RESUMEN

CX-5461 is currently in Phase I/II clinical trials for advanced hematologic malignancies and triple negative or BRCA-deficient breast cancer. The compound is currently administered to patients intravenously (i.v.) at low pH (3.5) due to solubility challenges. Reliance of low pH to enhance solubility of CX-5461 can adversely impact pharmacokinetics, biodistribution and therapeutic potential. We have addressed this solubility issue through a formulation method that relies on the interactions between CX-5461 and copper. Copper binds CX-5461 through the nitrogens of the pyrazine ring. Here, we describe synthesizing this copper-complexed CX-5461 (Cu(CX-5461)) within liposomes. CX-5461 was added to copper-containing liposomes and incubated at 60 °C for 30 min. The pharmacokinetics of CX-5461 was assessed in mice following a single i.v. injection at 30 mg/kg. Efficacy studies were completed in multiple subcutaneous mouse xenografts as well as in a bone marrow engraftment model of acute myeloid leukemia (AML). The novel Cu(CX-5461) formulation was stable at pH 7.4 and exhibited increased plasma circulation longevity, increasing the total exposure to CX5461 by an order of magnitude. Cu(CX-5461) was more active than CX-5461 in AML models in vivo. In HCT116-B46 and Capan-1 solid tumour models that are BRCA-deficient, the Cu(CX-5461) formulation engendered activity that was comparable to that of the low pH CX-5461 formulation. We have generated the first Cu(CX-5461) formulation suitable for i.v. administration that is more efficacious than the existing low-pH formulation in pre-clinical models of AML. The Cu(CX-5461) formulation may serve as an alternative formulation for CX-5461 in BRCA-deficient cancers.


Asunto(s)
Antineoplásicos/administración & dosificación , Benzotiazoles/administración & dosificación , Cobre/administración & dosificación , Naftiridinas/administración & dosificación , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Benzotiazoles/química , Benzotiazoles/farmacocinética , Benzotiazoles/uso terapéutico , Línea Celular Tumoral , Complejos de Coordinación/administración & dosificación , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Complejos de Coordinación/uso terapéutico , Cobre/química , Cobre/farmacocinética , Cobre/uso terapéutico , Femenino , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Liposomas/química , Ratones , Naftiridinas/química , Naftiridinas/farmacocinética , Naftiridinas/uso terapéutico , ARN Ribosómico/antagonistas & inhibidores , ARN Ribosómico/metabolismo , Distribución Tisular
17.
Oncol Rep ; 17(6): 1453-60, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17487404

RESUMEN

1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd, TAS-106), is a new antitumor cytidine analogue, inhibiting RNA synthesis. In this study we investigated the cellular growth inhibition, intracellular metabolism, cell cycle phase specificity, and RNA synthesis of TAS-106 compared with those of 5-fluorouracil (5-FU), known to possess both DNA- (inhibition of thymidylate synthase activity) and RNA-synthesis-inhibiting activity (inhibition of RNA function). The IC50 values of TAS-106 and 5-FU ranged from 0.0173 to 3.11 microM, and from 6.80 to >1,000 microM, respectively, in a panel of 10 human tumor cells, indicating that TAS-106 possesses greater cytotoxicity than 5-FU. Using excess thymidine-synchronized cells, TAS-106 and 5-FU appeared to exert their cytotoxic effects independently of the cell cycle. The intracellular metabolism and the effect on pre-rRNA processing of TAS-106 differed from those of 5-FU. More than 50% of 5-FU incorporated into the cells was in the unchanged form, while 5-FU incorporated into RNA was approximately 20%. On the other hand, TAS-106 was incorporated in a time-dependent manner into the cells and rapidly converted to its mono-, di- and tri-phosphate form, however, the amount incorporated into RNA fraction was very small. 5-FU incorporated into RNA was confirmed to impair the normal processing of ribosomal RNA (formation of 34/32S RNA from 45S RNA), however, TAS-106 did not affect pre-rRNA processing and may be involved in the inhibition of the synthesis of ribosomal RNA. We concluded that intracellular accumulation and retention of the active metabolite of TAS-106, 3'-ethynylcytidine 5'-triphosphate (ECTP), may contribute to its potent cytotoxicity. The unique mechanism of antitumor activity and intensive cellular metabolism of TAS-106 could contribute to cancer chemotherapy through the pathways different from those of 5-FU or other antitumor nucleosides.


Asunto(s)
Antineoplásicos/farmacología , Citidina/análogos & derivados , ARN Ribosómico/antagonistas & inhibidores , Ribonucleósidos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citidina/análisis , Citidina/metabolismo , Citidina/farmacología , Fluorouracilo/análisis , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Humanos , Concentración 50 Inhibidora , Precursores del ARN/antagonistas & inhibidores , Precursores del ARN/química , Precursores del ARN/metabolismo , ARN Ribosómico/química , ARN Ribosómico/metabolismo
18.
PLoS One ; 12(11): e0187977, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29190286

RESUMEN

The nucleolus produces the large polycistronic transcript (47S precursor) containing the 18S, 5.8S and 28S rRNA sequences and hosts most of the nuclear steps of pre-rRNA processing. Among numerous components it contains condensed chromatin and active rRNA genes which adopt a more accessible conformation. For this reason, it is a paradigm of chromosome territory organization. Active rRNA genes are clustered within several fibrillar centers (FCs), in which they are maintained in an open configuration by Upstream Binding Factor (UBF) molecules. Here, we used the reproducible reorganization of nucleolar components induced by the inhibition of rRNA synthesis by Actinomycin D (AMD) to address the steps of the spatiotemporal reorganization of FCs and nucleolar condensed chromatin. To reach that goal, we used two complementary approaches: i) time-lapse confocal imaging of cells expressing one or several GFP-tagged proteins (fibrillarin, UBF, histone H2B) and ii) ultrastructural identification of nucleolar components involved in the reorganization. Data obtained by time lapse confocal microscopy were analyzed through detailed 3D imaging. This allowed us to demonstrate that AMD treatment induces no fusion and no change in the relative position of the different nucleoli contained in one nucleus. In contrast, for each nucleolus, we observed step by step gathering and fusion of both FCs and nucleolar condensed chromatin. To analyze the reorganization of FCs and condensed chromatin at a higher resolution, we performed correlative light and electron microscopy electron microscopy (CLEM) imaging of the same cells. We demonstrated that threads of intranucleolar condensed chromatin are localized in a complex 3D network of vacuoles. Upon AMD treatment, these structures coalesce before migrating toward the perinucleolar condensed chromatin, to which they finally fuse. During their migration, FCs, which are all linked to ICC, are pulled by the latter to gather as caps disposed at the periphery of nucleoli.


Asunto(s)
Compartimento Celular , Nucléolo Celular/metabolismo , Cromatina/metabolismo , ARN Ribosómico/antagonistas & inhibidores , Línea Celular Tumoral , Dactinomicina/farmacología , Humanos , Microscopía Electrónica de Transmisión , ARN Ribosómico/biosíntesis , ARN Ribosómico/efectos de los fármacos
19.
ACS Infect Dis ; 3(3): 206-215, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28103015

RESUMEN

The antibacterial effects of aminoglycosides are based on their association with the A-site of bacterial rRNA and interference with the translational process in the bacterial cell, causing cell death. The clinical use of aminoglycosides is complicated by resistance and side effects, some of which arise from their interactions with the human mitochondrial 12S rRNA and its deafness-associated mutations, C1494U and A1555G. We report a rapid assay that allows screening of aminoglycoside compounds to these classes of rRNAs. These screening tools are important to find antibiotics that selectively bind to the bacterial A-site rather than human, mitochondrial A-sites and its mutant homologues. Herein, we report our preliminary work on the optimization of this screen using 12 anthraquinone-neomycin (AMA-NEO) conjugates against molecular constructs representing five A-site homologues, Escherichia coli, human cytosolic, mitochondrial, C1494U, and A1555G, using a fluorescent displacement screening assay. These conjugates were also tested for inhibition of protein synthesis, antibacterial activity against 14 clinically relevant bacterial strains, and the effect on enzymes that inactivate aminoglycosides. The AMA-NEO conjugates demonstrated significantly improved resistance against aminoglycoside-modifying enzymes (AMEs), as compared with NEO. Several compounds exhibited significantly greater inhibition of prokaryotic protein synthesis as compared to NEO and were extremely poor inhibitors of eukaryotic translation. There was significant variation in antibacterial activity and MIC of selected compounds between bacterial strains, with Escherichia coli, Enteroccocus faecalis, Citrobacter freundii, Shigella flexneri, Serratia marcescens, Proteus mirabilis, Enterobacter cloacae, Staphylococcus epidermidis, and Listeria monocytogenes exhibiting moderate to high sensitivity (50-100% growth inhibition) whereas Acinetobacter baumannii, Pseudomonas aeruginosa, Klebsiellla pneumoniae, and MRSA strains expressed low sensitivity, as compared to the parent aminoglycoside NEO.


Asunto(s)
Aminoglicósidos/farmacología , Antiinfecciosos/farmacología , ARN Ribosómico/antagonistas & inhibidores , Aminoglicósidos/química , Antraquinonas/química , Antraquinonas/farmacología , Antiinfecciosos/química , Sitios de Unión , Farmacorresistencia Microbiana/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Mutación , Neomicina/química , Neomicina/farmacología , ARN Bacteriano/antagonistas & inhibidores , ARN Bacteriano/química , ARN Ribosómico/química , ARN Ribosómico/genética
20.
Biomaterials ; 138: 1-12, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28550752

RESUMEN

The rising antimicrobial resistance contributes to 25000 annual deaths in Europe. This threat to the public health can only be tackled if novel antimicrobials are developed, combined with a more precise use of the currently available antibiotics through the implementation of fast, specific, diagnostic methods. Nucleic acid mimics (NAMs) that are able to hybridize intracellular bacterial RNA have the potential to become such a new class of antimicrobials and additionally could serve as specific detection probes. However, an essential requirement is that these NAMs should be delivered into the bacterial cytoplasm, which is a particular challenge given the fact that they are charged macromolecules. We consider these delivery challenges in relation to the gastric pathogen Helicobacter pylori, the most frequent chronic infection worldwide. In particular, we evaluate if cationic fusogenic liposomes are suitable carriers to deliver NAMs across the gastric mucus barrier and the bacterial envelope. Our study shows that DOTAP-DOPE liposomes post-PEGylated with DSPE-PEG (DSPE Lpx) can indeed successfully deliver NAMs into Helicobacter pylori, while offering protection to the NAMs from binding and inactivation in gastric mucus isolated from pigs. DSPE Lpx thus offer exciting new possibilities for in vivo diagnosis and treatment of Helicobacter pylori infections.


Asunto(s)
Antiinfecciosos/administración & dosificación , Sistemas de Liberación de Medicamentos , Farmacorresistencia Microbiana , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/metabolismo , Moco/química , Oligonucleótidos Antisentido/administración & dosificación , ARN Bacteriano/antagonistas & inhibidores , ARN Ribosómico/antagonistas & inhibidores , Animales , Antiinfecciosos/síntesis química , Antiinfecciosos/metabolismo , Citoplasma/metabolismo , Ácidos Grasos Monoinsaturados/química , Colorantes Fluorescentes/química , Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Hibridación Fluorescente in Situ , Liposomas , Imitación Molecular , Moco/microbiología , Oligonucleótidos/administración & dosificación , Oligonucleótidos/síntesis química , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Oligonucleótidos Antisentido/síntesis química , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Compuestos de Amonio Cuaternario/química , ARN Bacteriano/genética , ARN Ribosómico/genética , Estómago/microbiología , Porcinos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda