Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Immunity ; 45(4): 761-773, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27692612

RESUMEN

Imiquimod is a small-molecule ligand of Toll-like receptor-7 (TLR7) that is licensed for the treatment of viral infections and cancers of the skin. Imiquimod has TLR7-independent activities that are mechanistically unexplained, including NLRP3 inflammasome activation in myeloid cells and apoptosis induction in cancer cells. We investigated the mechanism of inflammasome activation by imiquimod and the related molecule CL097 and determined that K+ efflux was dispensable for NLRP3 activation by these compounds. Imiquimod and CL097 inhibited the quinone oxidoreductases NQO2 and mitochondrial Complex I. This induced a burst of reactive oxygen species (ROS) and thiol oxidation, and led to NLRP3 activation via NEK7, a recently identified component of this inflammasome. Metabolic consequences of Complex I inhibition and endolysosomal effects of imiquimod might also contribute to NLRP3 activation. Our results reveal a K+ efflux-independent mechanism for NLRP3 activation and identify targets of imiquimod that might be clinically relevant.


Asunto(s)
Inflamasomas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Potasio/metabolismo , ARN Nuclear Pequeño/farmacología , Animales , Complejo I de Transporte de Electrón/metabolismo , Ratones , Quinasas Relacionadas con NIMA/metabolismo , Quinona Reductasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 7/metabolismo
2.
J Neuroinflammation ; 20(1): 152, 2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37365617

RESUMEN

BACKGROUND: Ambient RNAs contamination in single-nuclei RNA sequencing (snRNA-seq) is a challenging problem, but the consequences of ambient RNAs contamination of damaged and/or diseased tissues are poorly understood. Cognitive impairments and white/gray matter injuries are characteristic of deeper cerebral hypoperfusion mouse models induced by bilateral carotid artery stenosis (BCAS), but the molecular mechanisms still need to be further explored. More importantly, the BCAS mice can also offer an excellent model to examine the signatures of ambient RNAs contamination in damaged tissues when performing snRNA-seq. METHODS: After the sham and BCAS mice were established, cortex-specific single-nuclei libraries were constructed. Single-nuclei transcriptomes were described informatically by the R package Seurat, and ambient RNA markers of were identified in each library. Then, after removing ambient RNAs in each sample using the in silico approaches, the combination of CellBender and subcluster cleaning, single-nuclei transcriptomes were reconstructed. Next, the comparison of ambient RNA contamination was performed using irGSEA analysis before and after the in silico approaches. Finally, further bioinformatic analyses were performed. RESULTS: The ambient RNAs are more predominant in the BCAS group than the sham group. The contamination mainly originated from damaged neuronal nuclei, but could be reduced largely using the in silico approaches. The integrative analysis of cortex-specific snRNA-seq data and the published bulk transcriptome revealed that microglia and other immune cells were the primary effectors. In the sequential microglia/immune subgroups analysis, the subgroup of Apoe+ MG/Mac (microglia/macrophages) was identified. Interestingly, this subgroup mainly participated in the pathways of lipid metabolism, associated with the phagocytosis of cell debris. CONCLUSIONS: Taken together, our current study unravels the features of ambient RNAs in snRNA-seq datasets under diseased conditions, and the in silico approaches can effectively eliminate the incorrected cell annotation and following misleading analysis. In the future, snRNA-seq data analysis should be carefully revisited, and ambient RNAs removal needs to be taken into consideration, especially for those diseased tissues. To our best knowledge, our study also offers the first cortex-specific snRNA-seq data of deeper cerebral hypoperfusion, which provides with novel therapeutic targets.


Asunto(s)
Estenosis Carotídea , Microglía , Animales , Ratones , Microglía/metabolismo , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/farmacología , ARN Nuclear Pequeño/uso terapéutico , Macrófagos , Estenosis Carotídea/complicaciones , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Ratones Endogámicos C57BL
3.
Toxicol Appl Pharmacol ; 471: 116550, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37172768

RESUMEN

The persistent environmental aryl hydrocarbon receptor agonist and hepatotoxin TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) induces hepatic lipid accumulation (steatosis), inflammation (steatohepatitis) and fibrosis. Thousands of liver-expressed, nuclear-localized lncRNAs with regulatory potential have been identified; however, their roles in TCDD-induced hepatoxicity and liver disease are unknown. We analyzed single nucleus (sn)RNA-seq data from control and subchronic (4 wk) TCDD-exposed mouse liver to determine liver cell-type specificity, zonation and differential expression profiles for thousands of lncRNAs. TCDD dysregulated >4000 of these lncRNAs in one or more liver cell types, including 684 lncRNAs specifically dysregulated in liver non-parenchymal cells. Trajectory inference analysis revealed major disruption by TCDD of hepatocyte zonation, affecting >800 genes, including 121 lncRNAs, with strong enrichment for lipid metabolism genes. TCDD also dysregulated expression of >200 transcription factors, including 19 Nuclear Receptors, most notably in hepatocytes and Kupffer cells. TCDD-induced changes in cell-cell communication patterns included marked decreases in EGF signaling from hepatocytes to non-parenchymal cells and increases in extracellular matrix-receptor interactions central to liver fibrosis. Gene regulatory networks constructed from the snRNA-seq data identified TCDD-exposed liver network-essential lncRNA regulators linked to functions such as fatty acid metabolic process, peroxisome and xenobiotic metabolism. Networks were validated by the striking enrichments that predicted regulatory lncRNAs showed for specific biological pathways. These findings highlight the power of snRNA-seq to discover functional roles for many xenobiotic-responsive lncRNAs in both hepatocytes and liver non-parenchymal cells and to elucidate novel aspects of foreign chemical-induced hepatotoxicity and liver disease, including dysregulation of intercellular communication within the liver lobule.


Asunto(s)
Hígado Graso , Hepatopatías , Dibenzodioxinas Policloradas , ARN Largo no Codificante , Ratones , Animales , Dibenzodioxinas Policloradas/toxicidad , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Xenobióticos/metabolismo , Hígado , Hígado Graso/metabolismo , Hepatopatías/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Comunicación Celular , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/farmacología
4.
Hum Mutat ; 40(8): 1172-1180, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31033086

RESUMEN

One of most important factors for messenger RNA (mRNA) transcription is the spliceosomal component U1 small nuclear RNA (snRNA), which recognizes 5' splicing donor sites at specific regions in pre-mRNA. Mutations in these sites disrupt U1 snRNA binding and cause abnormal splicing. In this study, we investigated mutations at splice sites in SLC26A4 (HGNC 8818), one of the major causative genes of hearing loss, which may result in the synthesis of abnormal pendrin, the channel protein encoded by the gene. Seventeen SLC26A4 variants with mutations in the U1 snRNA binding sites were assessed by minigene splicing assays, and 11 were found to result in abnormal splicing. Interestingly, eight of the 11 pathogenic mutations were intronic, suggesting the importance of conserved sequences at the intronic splice site. The application of modified U1 snRNA effectively rescued the abnormal splicing for most of these mutations. Although three were cryptic mutations, they were rescued by cotransfection of modified U1 snRNA and modified antisense oligonucleotides. Our results demonstrate the important role of snRNA in SLC26A4 mutations, suggesting the therapeutic potential of modified U1 snRNA and antisense oligonucleotides for neutralizing the pathogenic effect of the splice-site mutations that may result in hearing loss.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Oligonucleótidos Antisentido/farmacología , ARN Nuclear Pequeño/farmacología , Transportadores de Sulfato/genética , Empalme Alternativo/efectos de los fármacos , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Células HeLa , Pérdida Auditiva Sensorineural/terapia , Humanos , Intrones , Mutación , Sitios de Empalme de ARN , ARN Nuclear Pequeño/metabolismo , Transportadores de Sulfato/química , Transportadores de Sulfato/metabolismo
5.
Mol Ther ; 24(10): 1797-1805, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27456062

RESUMEN

Spinal Muscular Atrophy is due to the loss of SMN1 gene function. The duplicate gene SMN2 produces some, but not enough, SMN protein because most transcripts lack exon 7. Thus, promoting the inclusion of this exon is a therapeutic option. We show that a somatic gene therapy using the gene for a modified U7 RNA which stimulates this splicing has a profound and persistent therapeutic effect on the phenotype of a severe Spinal Muscular Atrophy mouse model. To this end, the U7 gene and vector and the production of pure, highly concentrated self-complementary (sc) adenovirus-associated virus 9 vector particles were optimized. Introduction of the functional vector into motoneurons of newborn Spinal Muscular Atrophy mice by intracerebroventricular injection led to a highly significant, dose-dependent increase in life span and improvement of muscle functions. Besides the central nervous system, the therapeutic U7 RNA was expressed in the heart and liver which may additionally have contributed to the observed therapeutic efficacy. This approach provides an additional therapeutic option for Spinal Muscular Atrophy and could also be adapted to treat other diseases of the central nervous system with regulatory small RNA genes.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Atrofia Muscular Espinal/terapia , ARN Nuclear Pequeño/administración & dosificación , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Animales , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Hígado/metabolismo , Ratones , Ratones Transgénicos , Atrofia Muscular Espinal/genética , Miocardio/metabolismo , Empalme del ARN , ARN Nuclear Pequeño/farmacología
6.
Brain ; 138(Pt 10): 2834-46, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26122121

RESUMEN

Defects of mitochondrial dynamics are emerging causes of neurological disease. In two children presenting with severe neurological deterioration following viral infection we identified a novel homozygous STAT2 mutation, c.1836 C>A (p.Cys612Ter), using whole exome sequencing. In muscle and fibroblasts from these patients, and a third unrelated STAT2-deficient patient, we observed extremely elongated mitochondria. Western blot analysis revealed absence of the STAT2 protein and that the mitochondrial fission protein DRP1 (encoded by DNM1L) is inactive, as shown by its phosphorylation state. All three patients harboured decreased levels of DRP1 phosphorylated at serine residue 616 (P-DRP1(S616)), a post-translational modification known to activate DRP1, and increased levels of DRP1 phosphorylated at serine 637 (P-DRP1(S637)), associated with the inactive state of the DRP1 GTPase. Knockdown of STAT2 in SHSY5Y cells recapitulated the fission defect, with elongated mitochondria and decreased P-DRP1(S616) levels. Furthermore the mitochondrial fission defect in patient fibroblasts was rescued following lentiviral transduction with wild-type STAT2 in all three patients, with normalization of mitochondrial length and increased P-DRP1(S616) levels. Taken together, these findings implicate STAT2 as a novel regulator of DRP1 phosphorylation at serine 616, and thus of mitochondrial fission, and suggest that there are interactions between immunity and mitochondria. This is the first study to link the innate immune system to mitochondrial dynamics and morphology. We hypothesize that variability in JAK-STAT signalling may contribute to the phenotypic heterogeneity of mitochondrial disease, and may explain why some patients with underlying mitochondrial disease decompensate after seemingly trivial viral infections. Modulating JAK-STAT activity may represent a novel therapeutic avenue for mitochondrial diseases, which remain largely untreatable. This may also be relevant for more common neurodegenerative diseases, including Alzheimer's, Huntington's and Parkinson's diseases, in which abnormalities of mitochondrial morphology have been implicated in disease pathogenesis.


Asunto(s)
Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Dinámicas Mitocondriales/fisiología , Factor de Transcripción STAT2/deficiencia , Transducción de Señal/genética , Apoptosis/genética , Preescolar , Dinaminas , Electroencefalografía , Salud de la Familia , Femenino , Citometría de Flujo , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Humanos , Lactante , Masculino , Microscopía Electrónica , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Neuroblastoma/patología , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Nuclear Pequeño/farmacología , Factor de Transcripción STAT2/genética , Transfección
7.
Nat Cell Biol ; 3(1): 97-9, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11146633

RESUMEN

All major nuclear export pathways so far examined follow a general paradigm. Specifically, a complex is formed in the nucleus, containing the export cargo, a member of the importin-beta family of transporters and RanGTP. This complex is translocated across the nuclear pore to the cytoplasm, where hydrolysis of the GTP on Ran is stimulated by the GTPase-activating protein RanGAP. The activity of RanGAP is increased by RanBP1, which also promotes disassembly of RanGTP-cargo-transporter complexes. Here we investigate the role of RanGTP in the export of mRNAs generated by splicing. We show that nuclear injection of a Ran mutant (RanT24N) or the normally cytoplasmic RanGAP potently inhibits the export of both tRNA and U1 snRNA, but not of spliced mRNAs. Moreover, nuclear injection of RanGAP together with RanBP1 blocks tRNA export but does not affect mRNA export. These and other data indicate that export of spliced mRNA is the first major cellular transport pathway that is independent of the export co-factor Ran.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Núcleo Celular/metabolismo , Empalme del ARN/fisiología , ARN Mensajero/metabolismo , Proteína de Unión al GTP ran/metabolismo , Animales , Núcleo Celular/ultraestructura , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Activadoras de GTPasa/farmacología , Cinética , Mutación/fisiología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , ARN Nuclear Pequeño/metabolismo , ARN Nuclear Pequeño/farmacología , ARN de Transferencia/metabolismo , ARN de Transferencia/farmacología , Xenopus , Proteínas de Xenopus
8.
ChemMedChem ; 16(20): 3181-3184, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34233081

RESUMEN

The 332-nucleotide small nuclear RNA (snRNA) 7SK is a highly conserved non-coding RNA that regulates transcriptional elongation. By binding with positive transcriptional elongation factor b (P-TEFb) via HEXIM1, 7SK snRNA decreases the kinase activity of P-TEFb and inhibits transcriptional elongation. Additionally, it is reported that 7SK inhibition results in the stimulation of human immunodeficiency virus (HIV)-specific transcription. These reports suggest that 7SK is a naturally occurring functional molecule as negative regulator of P-TEFb and HIV transcription. In this study, we developed functional oligonucleotides that mimic the function of 7SK (7SK mimics) as novel inhibitors of HIV replication. We defined the essential region of 7SK regarding its suppressive effects on transcriptional downregulation using an antisense strategy. Based on the results, we designed 7SK mimics containing the defined region. The inhibitory effects of 7SK mimics on HIV-1 long terminal repeat promoter specific transcription was drastic compared with those of the control mimic molecule. Notably, these effects were found to be more enhanced by co-transfection with Tat-expressing plasmids. From these results, it is indicated that 7SK mimics may have great therapeutic potential for HIV/AIDS treatment.


Asunto(s)
Desarrollo de Medicamentos , ARN Nuclear Pequeño/farmacología , Transcripción Genética/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Estructura Molecular , ARN Nuclear Pequeño/síntesis química , ARN Nuclear Pequeño/química , Relación Estructura-Actividad , Transcripción Genética/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
9.
Mini Rev Med Chem ; 9(3): 379-85, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19275730

RESUMEN

Transcription is considered to be a crucial step in the replication cycle of HIV-1. Tat regulates an early step of transcription elongation. The positive elongation factor P-TEFb, a heterodimer containing a catalytic subunit (CDK9) and unique regulatory cyclins (CycT1), is required for HIV-1 Tat transcriptional activation. This is a potential target for new HIV-1 transcription inhibitors. Without P-TEFb, transactivation is restrained and only short transcripts are generated. All the P-TEFb inhibitors can suppress the HIV-1 transactivation process by inhibition of CycT1, CDK9 or their interaction. Several low-molecular-weight compounds such as flavopiridol, roscovitine and the human small nuclear RNA 7SK which have been showed to possess potent anti-HIV activity by interfering with P-TEFb functions are reviewed in this article.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , VIH-1/fisiología , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/fisiología , Flavonoides/química , Flavonoides/farmacología , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Humanos , Modelos Biológicos , Piperidinas/química , Piperidinas/farmacología , Factor B de Elongación Transcripcional Positiva/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Purinas/química , Purinas/farmacología , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/farmacología , Roscovitina , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología
10.
Nucleic Acids Res ; 35(1): 247-55, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17158512

RESUMEN

Control of RNA processing plays a central role in regulating the replication of HIV-1, in particular the 3' polyadenylation of viral RNA. Based on the demonstration that polyadenylation of mRNAs can be disrupted by the targeted binding of modified U1 snRNA, we examined whether binding of U1 snRNAs to conserved 10 nt regions within the terminal exon of HIV-1 was able to inhibit viral structural protein expression. In this report, we demonstrate that U1 snRNAs complementary to 5 of the 15 regions targeted result in significant suppression of HIV-1 protein expression and viral replication coincident with loss of viral RNA. Suppression of viral gene expression is dependent upon appropriate assembly of a U1 snRNP particle as mutations of U1 snRNA that affect binding of U1 70K or Sm proteins significantly reduced efficacy. However, constructs lacking U1A binding sites retained significant anti-viral activity. This finding suggests a role for these mutants in situations where the wild-type constructs cause toxic effects. The conserved nature of the sequences targeted and the high efficacy of the constructs suggests that this strategy has significant potential as an HIV therapeutic.


Asunto(s)
Fármacos Anti-VIH/química , VIH-1/efectos de los fármacos , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/farmacología , Replicación Viral/efectos de los fármacos , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/farmacología , Secuencia de Bases , Línea Celular , Biología Computacional , Exones , Regulación Viral de la Expresión Génica , VIH-1/genética , VIH-1/fisiología , Humanos , Datos de Secuencia Molecular , Mutación , ARN Mensajero/química , ARN Nuclear Pequeño/química , ARN Viral/química , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Replicación Viral/genética
11.
Cell Mol Gastroenterol Hepatol ; 8(3): 407-426, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31195149

RESUMEN

BACKGROUND & AIMS: The small intestine (SI) displays regionality in nutrient and immunological function. Following SI tissue loss (as occurs in short gut syndrome, or SGS), remaining SI must compensate, or "adapt"; the capacity of SI epithelium to reprogram its regional identity has not been described. Here, we apply single-cell resolution analyses to characterize molecular changes underpinning adaptation to SGS. METHODS: Single-cell RNA sequencing was performed on epithelial cells isolated from distal SI of mice following 50% proximal small bowel resection (SBR) vs sham surgery. Single-cell profiles were clustered based on transcriptional similarity, reconstructing differentiation events from intestinal stem cells (ISCs) through to mature enterocytes. An unsupervised computational approach to score cell identity was used to quantify changes in regional (proximal vs distal) SI identity, validated using immunofluorescence, immunohistochemistry, qPCR, western blotting, and RNA-FISH. RESULTS: Uniform Manifold Approximation and Projection-based clustering and visualization revealed differentiation trajectories from ISCs to mature enterocytes in sham and SBR. Cell identity scoring demonstrated segregation of enterocytes by regional SI identity: SBR enterocytes assumed more mature proximal identities. This was associated with significant upregulation of lipid metabolism and oxidative stress gene expression, which was validated via orthogonal analyses. Observed upstream transcriptional changes suggest retinoid metabolism and proximal transcription factor Creb3l3 drive proximalization of cell identity in response to SBR. CONCLUSIONS: Adaptation to proximal SBR involves regional reprogramming of ileal enterocytes toward a proximal identity. Interventions bolstering the endogenous reprogramming capacity of SI enterocytes-conceivably by engaging the retinoid metabolism pathway-merit further investigation, as they may increase enteral feeding tolerance, and obviate intestinal failure, in SGS.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Intestino Delgado/cirugía , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Reprogramación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Enterocitos/química , Enterocitos/citología , Intestino Delgado/química , Metabolismo de los Lípidos , Masculino , Ratones , Estrés Oxidativo , ARN Nuclear Pequeño/farmacología , Aprendizaje Automático no Supervisado , Regulación hacia Arriba
12.
J Mol Biol ; 286(5): 1347-63, 1999 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-10064702

RESUMEN

A molecular dissection of U3 small nucleolar RNA (snoRNA) was performed in vivo in Xenopus oocytes and the effects on rRNA processing were analyzed. Oocyte injection of antisense oligonucleotides against parts of U3 snoRNA resulted in specific fragmentation of U3 by endogenous RNase H. Fragmentation of U3 domain II correlated with a decrease in 20 S pre-rRNA and a concomitant increase in 36 S pre-rRNA, indicating reduced cleavage at site 3. Conversely, fragmentation of U3 domain I completely blocked 18 S rRNA formation, increased the 20 S rRNA precursor, and decreased 36 S pre-rRNA, indicating inhibition of cleavage at sites 1+2. rRNA processing defects at sites 1+2 or 3 after destruction of intact endogenous U3 snoRNA were rescued by injection of in vitro transcripts of U3 snoRNA or certain U3 fragments. Thus, cleavage at sites 1+2 and 3 is U3 snoRNA dependent. Moreover, U3 snoRNA has two functional modules: domain I for sites 1+2 cleavage and domain II for site 3 cleavage. The data suggest that whichever of these U3 domains acts first determines which rRNA processing pathway will be taken: cleavage first at site 3 of pre-rRNA leads to pathway A, whereas cleavage first at sites 1+2 leads to pathway B for rRNA processing. Predictions of this model were validated by rescue of site 3 cleavage by injection of just domain II after U3 depletion. Rescue of sites 1+2 cleavage required covalent continuity of domain I with the hinge region and non-covalent association with domain II. We could experimentally shift which rRNA processing pathway was taken by injecting fragments of U3 to compete with endogenous U3 snoRNA.


Asunto(s)
Nucléolo Celular/metabolismo , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Ribosómico/metabolismo , ARN Nuclear Pequeño/metabolismo , Animales , Secuencia de Bases , Northern Blotting , Nucléolo Celular/genética , Datos de Secuencia Molecular , Peso Molecular , Conformación de Ácido Nucleico/efectos de los fármacos , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Oligodesoxirribonucleótidos Antisentido/genética , Oligodesoxirribonucleótidos Antisentido/farmacología , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Ribosómico/genética , ARN Ribosómico 18S/genética , ARN Ribosómico 18S/metabolismo , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/farmacología , Ribonucleasa H/metabolismo , Factores de Tiempo , Xenopus laevis
13.
FEBS Lett ; 491(3): 200-6, 2001 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-11240127

RESUMEN

Previous studies indicated that transforming RNA, derived from the 3' half of the U5 small nuclear RNA first stem structure, suppressed the secretory protein translation in vitro. Gap junctions facilitate homeostatic control of cell growth and differentiation and their dysfunction has been correlated with carcinogenesis. Here, we reported that transforming RNA directly suppressed the gap junction protein, connexin 43, translation and thereby inhibited functional gap junction function in rat epithelial cells. Together with previous data, this implies that altered expression of transforming RNA itself is a potential mechanism in inhibiting gap junction function during carcinogenesis.


Asunto(s)
Comunicación Celular/fisiología , Células Epiteliales/metabolismo , Uniones Comunicantes/metabolismo , Hígado/metabolismo , ARN Nuclear Pequeño/metabolismo , Animales , Western Blotting , Comunicación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Conexina 43/antagonistas & inhibidores , Conexina 43/biosíntesis , Conexina 43/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Uniones Comunicantes/efectos de los fármacos , Hígado/citología , Conformación de Ácido Nucleico , Biosíntesis de Proteínas/efectos de los fármacos , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/farmacología , Ratas , Transfección
14.
World J Gastroenterol ; 9(3): 572-7, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12632521

RESUMEN

AIM: To study the preparation and cleavage activity of anti-transforming growth factor (TGF)beta1 U1 small nuclear (sn) RNA chimeric hammerhead ribozymes in vitro. METHODS: TGFbeta1 partial gene fragment was cloned into T-vector at the downstream of T7 promoter. (32)p-labeled TGFbeta1 partial transcripts as target RNA were transcribed in vitro and purified by denaturing polyacrylamide gel electrophoresis (PAGE). Anti-TGFbeta1 ribozymes were designed by computer, then synthetic ribozyme fragments were cloned into the U1 ribozyme vector pZeoU1EcoSpe containing U1 snRNA promoter/enhancer and terminator. (32)p-labeled U1 snRNA chimeric ribozyme transcripts were gel-purified, incubated with target-RNAs at different conditions and autoradiographed after running denaturing PAGE. RESULTS: Active U1snRNA chimeric ribozyme (U1Rz803) had the best cleavage activity at 50 degrees; at 37 degrees, it was active, K(m)=34.48 nmol/L, K(cat)=0.14 min(-1); while the point mutant ribozyme U1Rz803(m) had no cleavage activity, so these indicated the design of U1Rz803 was correct. CONCLUSION: U1Rz803 prepared in this study possessed the perfect specific catalytic cleavage activity. These results indicate U1 snRNA chimeric ribozyme U1Rz803 may suppress the expression of TGFbeta1 in vivo, therefore it may provide a new avenue for the treatment of liver fibrosis in the future.


Asunto(s)
ARN Catalítico/farmacología , ARN Nuclear Pequeño/farmacología , ARN/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética , Animales , Línea Celular Transformada , Clonación Molecular , Ratas , Factor de Crecimiento Transformador beta1
15.
Mutat Res ; 326(1): 71-82, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7528887

RESUMEN

U5 snRNA can induce both transformation and chromosome aberrations of cells. The polypurine tract, GGAGAGGAA, of the RNA has been suggested to participate in both phenomena. In vitro transcription expected to give this polypurine oligoribonucleotide was associated with cleavage of transcripts, generating 5'-terminal hydroxyl and 3'-terminal 2',3'-cyclic phosphate groups. The cleavage was further studied by making use of a Mg(2+)-catalyzed reaction and RNase T1 and RNase U2 digestion. The cleavage was found to generate highly reactive RNA molecules, participating in subsequent ligation of RNAs. Such a reactive molecule, guanosine-2',3'-cyclic phosphate, was capable of blocking DNA synthesis in vitro. The results may provide a possible mechanism of the chromosome aberrations induced by U5.


Asunto(s)
Aberraciones Cromosómicas , ADN/efectos de los fármacos , Nucleótidos de Guanina/farmacología , ARN Nuclear Pequeño/farmacología , Secuencia de Bases , Catálisis , ADN/biosíntesis , Magnesio/metabolismo , Datos de Secuencia Molecular , ARN/metabolismo , Moldes Genéticos , Transcripción Genética
16.
Zhonghua Gan Zang Bing Za Zhi ; 12(12): 749-51, 2004 Dec.
Artículo en Zh | MEDLINE | ID: mdl-15619345

RESUMEN

OBJECTIVE: To study the cleavage activity on the HCV RNA of a chimeric recombinant of HCV specific ribozyme and U1 small nuclear RNA, which compartmentalizes within the nucleolus. METHODS: The third stem-loop sequence of human U1 snRNA (position 95-116) within pBSIISK+ U1 was substituted by hammerhead ribozyme against HCV RNA by PCR and cloning methods, and the constructed plasmid was named pBSIISK+ (U1-Rz). Then the whole gene fragment of the chimeric ribozyme was cloned into a pGEM-T vector under the control of T7 promoter, and the constructed plasmid was named pGEM- (U1-Rz). The pGEM- (U1-Rz) and pGEM-Rz (containing the same ribozyme sequence as that in U1-Rz) transcripts as enzyme were transcribed in vitro. Also the (32)P-labeled pCMV/T7-NCRC luc (containing the gene sequence of the whole 5'-NCR and part core of HCV RNA) transcripts as target-RNAs were transcribed in vitro. The enzymes were incubated with the target RNAs under different conditions and autoradiographed after denaturing gel-electrophoresis. RESULTS: The sequencing result showed that the construction of U1 snRNA chimeric ribozyme was correct. Compared with the ribozyme alone, both of them were active at 37 degree C and with Mg2+ (10 mmol/L) and TrisCl (10 mmol/L, pH7.9), and there was no remarkable difference between them. The cleavage activity of the chimeric ribozyme increased with the prolongation of reaction time and increment of enzyme concentration. CONCLUSION: Both ribozyme and U1 snRNA chimeric ribozyme exhibited specifically catalytic activity against HCV RNA in vitro. There was no remarkable difference between their cleavage efficiencies.


Asunto(s)
Hepacivirus/genética , ARN Catalítico/genética , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/farmacología , Proteínas Recombinantes de Fusión/farmacología , Quimera/genética , Terapia Genética , Hepatitis C/terapia , ARN Catalítico/metabolismo , ARN Viral/genética
18.
Neurotox Res ; 16(3): 261-70, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19526279

RESUMEN

The canonical histopathological feature of Parkinson's disease (PD) is the loss of dopaminergic neurons in the ventral midbrain. Although the common sporadic/idiopathic form of PD most often presents clinically at around 60 years of age when the levels of striatal dopamine and numbers of ventral dopaminergic neurons are posited to have declined by 80 and 60%, respectively, the temporal pattern of injury to these vulnerable cells is unknown. The conventional view is that PD results from an accelerated age-related loss of dopamine neurons. However, an alternative hypothesis is that dopamine neuron loss is a developmental phenomenon. What evidence might support this alternative view? Apart from the rare familial forms, wherein loss or gain of function mutations in single genes convey highly penetrant PD, sporadic disease is genetically complex and may have other contributory non-genetic components. Epidemiologic and twin studies have strongly implicated gene-environmental interaction as a pathogenic dyad in the etiology of PD. Among the most attractive candidates that may connect the environment to inherited vulnerability is the nuclear receptor, Nurr1. Encoding an orphan transcription factor that is expressed at high levels within discrete regions of the developing and adult mammalian brain, Nurr1 is essential for the formation of ventral midbrain dopamine neurons. Given the absence of a known lipophilic small molecule regulator and established transcriptional role in the formation of the definitive dopaminergic phenotype, Nurr1 represents an intriguing molecule to explore in the context of sporadic PD as a developmental disorder. The study described herein addresses two features of Nurr1 biology that provide plausibility for this hypothesis. First is the description of Nurr1 regulation of a potent dopaminergic neuronal trophic factor, vasoactive intestinal peptide (VIP), and second is the identification of a protein, termed Nurr1 interacting protein (NuIP) that appears to link upstream signaling pathways in the regulation of Nurr1 transcriptional activity.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Modelos Biológicos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Factores de Transcripción/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Dopamina/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Neuronas/patología , Neuronas/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Enfermedad de Parkinson/patología , ARN Nuclear Pequeño/farmacología , Transducción de Señal/fisiología , Factores de Transcripción/genética , Péptido Intestinal Vasoactivo/metabolismo
19.
J Gene Med ; 9(5): 323-34, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17474072

RESUMEN

The HIV-1 regulatory proteins Tat and Rev are encoded by multiply spliced mRNAs that differ by the use of alternative 3' splice sites at the beginning of the internal exon. If these internal exons are skipped, the expression of these genes, and hence HIV-1 multiplication, should be inhibited. We have previously developed a strategy, based on antisense derivatives of U7 small nuclear RNA, that allows us to induce the skipping of an internal exon in virtually any gene. Here, we have successfully applied this approach to induce a partial skipping of the Tat, Rev (and Nef) internal exons. Three functional U7 constructs were subcloned into a lentiviral vector. Two of them strongly reduced the efficiency of lentiviral particle production compared to vectors carrying either no U7 insert or unrelated U7 cassettes. This defect could be partly or fully compensated by coexpressing Rev from an unspliced mRNA in the producing cell line. Upon stable transduction into CEM-SS or CEM T-lymphocytes, the most efficient of these constructs inhibits HIV-1 multiplication. Although the inhibition is not complete, it is more efficient in combination with another mechanism inhibiting HIV multiplication. Therefore, this new approach targeting HIV-1 regulatory genes at the level of pre-mRNA splicing, in combination with other antiviral strategies, may be a useful new tool in the fight against HIV/AIDS.


Asunto(s)
Exones , Genes Reguladores/efectos de los fármacos , VIH-1/genética , ARN Nuclear Pequeño/farmacología , Replicación Viral/efectos de los fármacos , Línea Celular , Genes rev/efectos de los fármacos , Genes tat/efectos de los fármacos , Vectores Genéticos , Humanos , Empalme del ARN , Linfocitos T/virología , Transducción Genética , Replicación Viral/genética
20.
J Gene Med ; 7(7): 965-76, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15772939

RESUMEN

BACKGROUND/AIMS: Transforming growth factor beta (TGFbeta1) is considered the key mediator in the process of liver fibrosis. The purpose of this investigation was to evaluate the activity of ribozymes against TGFbeta1 in a cell-free system and activated hepatic stellate cells (HSCs), and antifibrotic effect in activated HSCs in vitro and in rats. METHODS: Three ribozymes targeting against TGFbeta1 mRNA were designed, and then cloned into the U1 snRNA expression cassette. The chimeric ribozymes were selected for the analysis of their performances in activated HSCs through the detection of their cleavage activities in a cell-free system. After ribozyme-encoding plasmids had been transfected into HSC-T6 cells, the effects of ribozymes on activated HSCs were evaluated through the analysis of proliferation, activation and collagen deposition of HSC-T6. The adenoviral vector expressing the ribozymes was constructed, and then delivered into rat models of hepatic fibrosis induced by carbon tetrachloride. RESULTS: TGFbeta1 expression was efficiently down-regulated in activated HSCs by U1 snRNA chimeric ribozymes which possessed perfect cleavage activity in a cell-free system. Further studies demonstrated that U1 snRNA chimeric ribozymes inhibited the synthesis of collagen I, reduced deposition of collagen I, suppressed BrdU incorporation, but had no effect on desmin and alpha-SMA expression in transfected HSC-T6 cells. Histological analysis demonstrated that the adenoviral vector expressing ribozyme (Rz803) could alleviate fibrotic pathology in rats treated with carbon tetrachloride. CONCLUSIONS: The anti-TGFbeta1 ribozymes could reverse the character of activated HSCs in vitro and improve fibrotic pathology in vivo. It indicated that TGFbeta1 could be considered as a novel candidate for a therapeutic agent against hepatic fibrosis.


Asunto(s)
Cirrosis Hepática/prevención & control , Hígado/citología , ARN Catalítico/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Secuencia de Bases , Proliferación Celular/efectos de los fármacos , Sistema Libre de Células , Células Cultivadas , Colágeno Tipo I/efectos de los fármacos , Colágeno Tipo I/metabolismo , Vectores Genéticos , Interferón gamma/metabolismo , Cirrosis Hepática/inducido químicamente , Modelos Biológicos , Datos de Secuencia Molecular , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/farmacología , Ratas , Transfección
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda