Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neuroinflammation ; 20(1): 152, 2023 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-37365617

RESUMO

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.


Assuntos
Estenose das Carótidas , Microglia , Animais , Camundongos , Microglia/metabolismo , RNA Nuclear Pequeno/metabolismo , RNA Nuclear Pequeno/farmacologia , RNA Nuclear Pequeno/uso terapêutico , Macrófagos , Estenose das Carótidas/complicações , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Camundongos Endogâmicos C57BL
2.
Toxicol Appl Pharmacol ; 471: 116550, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37172768

RESUMO

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.


Assuntos
Fígado Gorduroso , Hepatopatias , Dibenzodioxinas Policloradas , RNA Longo não Codificante , Camundongos , Animais , Dibenzodioxinas Policloradas/toxicidade , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Xenobióticos/metabolismo , Fígado , Fígado Gorduroso/metabolismo , Hepatopatias/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Comunicação Celular , RNA Nuclear Pequeno/metabolismo , RNA Nuclear Pequeno/farmacologia
3.
ChemMedChem ; 16(20): 3181-3184, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34233081

RESUMO

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.


Assuntos
Desenvolvimento de Medicamentos , RNA Nuclear Pequeno/farmacologia , Transcrição Gênica/efeitos dos fármacos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Relação Dose-Resposta a Droga , Estrutura Molecular , RNA Nuclear Pequeno/síntese química , RNA Nuclear Pequeno/química , Relação Estrutura-Atividade , Transcrição Gênica/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
4.
Cell Mol Gastroenterol Hepatol ; 8(3): 407-426, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31195149

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Intestino Delgado/cirurgia , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Animais , Reprogramação Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Enterócitos/química , Enterócitos/citologia , Intestino Delgado/química , Metabolismo dos Lipídeos , Masculino , Camundongos , Estresse Oxidativo , RNA Nuclear Pequeno/farmacologia , Aprendizado de Máquina não Supervisionado , Regulação para Cima
5.
Hum Mutat ; 40(8): 1172-1180, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31033086

RESUMO

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.


Assuntos
Perda Auditiva Neurossensorial/genética , Oligonucleotídeos Antissenso/farmacologia , RNA Nuclear Pequeno/farmacologia , Transportadores de Sulfato/genética , Processamento Alternativo/efeitos dos fármacos , Sequência de Bases , Sítios de Ligação , Sequência Conservada , Células HeLa , Perda Auditiva Neurossensorial/terapia , Humanos , Íntrons , Mutação , Sítios de Splice de RNA , RNA Nuclear Pequeno/metabolismo , Transportadores de Sulfato/química , Transportadores de Sulfato/metabolismo
6.
Immunity ; 45(4): 761-773, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27692612

RESUMO

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.


Assuntos
Inflamassomos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Potássio/metabolismo , RNA Nuclear Pequeno/farmacologia , Animais , Complexo I de Transporte de Elétrons/metabolismo , Camundongos , Quinases Relacionadas a NIMA/metabolismo , Quinona Redutases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor 7 Toll-Like/metabolismo
7.
Mol Ther ; 24(10): 1797-1805, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27456062

RESUMO

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.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Atrofia Muscular Espinal/terapia , RNA Nuclear Pequeno/administração & dosagem , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Animais , Modelos Animais de Doenças , Vetores Genéticos/administração & dosagem , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Atrofia Muscular Espinal/genética , Miocárdio/metabolismo , Splicing de RNA , RNA Nuclear Pequeno/farmacologia
8.
Brain ; 138(Pt 10): 2834-46, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26122121

RESUMO

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.


Assuntos
Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Dinâmica Mitocondrial/fisiologia , Fator de Transcrição STAT2/deficiência , Transdução de Sinais/genética , Apoptose/genética , Pré-Escolar , Dinaminas , Eletroencefalografia , Saúde da Família , Feminino , Citometria de Fluxo , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Células HEK293 , Humanos , Lactente , Masculino , Microscopia Eletrônica , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Neuroblastoma/patologia , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Nuclear Pequeno/farmacologia , Fator de Transcrição STAT2/genética , Transfecção
9.
Neurotox Res ; 16(3): 261-70, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19526279

RESUMO

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.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Modelos Biológicos , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Doença de Parkinson/etiologia , Doença de Parkinson/genética , Fatores de Transcrição/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Dopamina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Neurônios/patologia , Neurônios/fisiologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Doença de Parkinson/patologia , RNA Nuclear Pequeno/farmacologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Peptídeo Intestinal Vasoativo/metabolismo
10.
Mini Rev Med Chem ; 9(3): 379-85, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19275730

RESUMO

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.


Assuntos
Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Fator B de Elongação Transcricional Positiva/antagonistas & inibidores , Fator B de Elongação Transcricional Positiva/fisiologia , Flavonoides/química , Flavonoides/farmacologia , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Humanos , Modelos Biológicos , Piperidinas/química , Piperidinas/farmacologia , Fator B de Elongação Transcricional Positiva/química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Purinas/química , Purinas/farmacologia , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/farmacologia , Roscovitina , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
11.
J Gene Med ; 9(5): 323-34, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17474072

RESUMO

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.


Assuntos
Éxons , Genes Reguladores/efeitos dos fármacos , HIV-1/genética , RNA Nuclear Pequeno/farmacologia , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Genes rev/efeitos dos fármacos , Genes tat/efeitos dos fármacos , Vetores Genéticos , Humanos , Splicing de RNA , Linfócitos T/virologia , Transdução Genética , Replicação Viral/genética
12.
Nucleic Acids Res ; 35(1): 247-55, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17158512

RESUMO

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.


Assuntos
Fármacos Anti-HIV/química , HIV-1/efeitos dos fármacos , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/farmacologia , Replicação Viral/efeitos dos fármacos , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , Sequência de Bases , Linhagem Celular , Biologia Computacional , Éxons , Regulação Viral da Expressão Gênica , HIV-1/genética , HIV-1/fisiologia , Humanos , Dados de Sequência Molecular , Mutação , RNA Mensageiro/química , RNA Nuclear Pequeno/química , RNA Viral/química , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/genética , Replicação Viral/genética
13.
J Biol Chem ; 280(39): 33123-31, 2005 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16051608

RESUMO

Dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) is a cell surface C-type lectin expressed on myeloid dendritic cells and certain tissue macrophages, which mediates antigen capture for processing and presentation and participates in intercellular interactions with naive T lymphocytes or endothelial cells. In their strategy to evade immunosurveillance, numerous pathogenic microorganisms, including human immunodeficiency virus and Mycobacterium, bind to DC-SIGN in order to gain access to dendritic cells. We present evidence that PU.1 dictates the basal and cell-specific activity of DC-SIGN gene-regulatory region through in vivo occupancy of two functional Ets elements, whose integrity is required for PU.1 responsiveness and for the cooperative actions of PU.1 and other transcription factors (Myb, RUNX) on the DC-SIGN gene proximal regulatory region. In addition, protein analysis and gene profiling experiments indicate that DC-SIGN and PU.1 are coordinately expressed upon classical and alternative macrophage activation and during dendritic cell maturation. Moreover, small interfering RNA-mediated reduction of PU.1 expression results in diminished DC-SIGN cellular levels. Altogether, these results indicate that PU.1 is involved in the myeloid-specific expression of DC-SIGN in myeloid cells, a contribution that can be framed within the role that PU.1 has on the acquisition of the antigen uptake molecular repertoire by dendritic cells and macrophages.


Assuntos
Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transativadores/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Western Blotting , Células COS , Células Cultivadas , Chlorocebus aethiops , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Humanos , Interleucina-4/farmacologia , Células Jurkat , Células K562 , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Ativação de Macrófagos , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Mutagênese Sítio-Dirigida , Células NIH 3T3 , Proteínas Proto-Oncogênicas/genética , RNA Nuclear Pequeno/farmacologia , Transativadores/genética
15.
J Gene Med ; 7(7): 965-76, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15772939

RESUMO

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.


Assuntos
Cirrose Hepática/prevenção & controle , Fígado/citologia , RNA Catalítico/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Sequência de Bases , Proliferação de Células/efeitos dos fármacos , Sistema Livre de Células , Células Cultivadas , Colágeno Tipo I/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Vetores Genéticos , Interferon gama/metabolismo , Cirrose Hepática/induzido quimicamente , Modelos Biológicos , Dados de Sequência Molecular , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/farmacologia , Ratos , Transfecção
16.
Zhonghua Gan Zang Bing Za Zhi ; 12(12): 749-51, 2004 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-15619345

RESUMO

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.


Assuntos
Hepacivirus/genética , RNA Catalítico/genética , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Quimera/genética , Terapia Genética , Hepatite C/terapia , RNA Catalítico/metabolismo , RNA Viral/genética
17.
J Natl Cancer Inst ; 96(11): 862-72, 2004 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-15173270

RESUMO

BACKGROUND: RNA interference initiated by small interfering RNAs effectively suppresses gene expression, but the suppression is transient, which limits the therapeutic use of this technique. Polo-like kinase 1 (PLK1) is a key cell cycle regulator that is overexpressed in various human tumors. We used a xenograft mouse model to determine whether an RNA interference-based strategy that used short hairpin RNAs (shRNAs) to suppress PLK1 expression could inhibit tumor growth in vivo. METHODS: HeLa S3 cervical and A549 lung cancer cell lines were transfected with plasmids containing U6 promoter-driven shRNAs against human PLK1 or control (parental or scrambled) plasmids. Plasmids were treated with the nuclease inhibitor aurintricarboxylic acid (ATA) as protection against nucleases in murine blood. Nude mice carrying xenograft tumors were injected with shRNA plasmids, and their xenograft tumor growth was assessed. Northern and western blot analyses were used to measure PLK1 mRNA and protein expression, respectively, in transfected cultured cells and in xenograft tumors. All statistical tests were two-sided. RESULTS: Levels of PLK1 mRNA and protein were lower in HeLa S3 and A549 cancer cells transfected with PLK1 shRNA plasmids than in corresponding cells transfected with control parental or scrambled PLK1S shRNA plasmids. Proliferation of cells transfected with PLK1 shRNA was lower than that of cells transfected with either control plasmid, and proliferation of cells transfected with ATA-treated PLK1 shRNA plasmids was even lower. In mice with human xenograft tumors, PLK1 shRNA expression from ATA-treated plasmids reduced tumor growth to 18% (95% confidence interval [CI] = 12% to 26%; P =.03) and from untreated plasmids reduced tumor growth to 45% (95% CI = 26% to 64%; P =.1) of that of tumors in mice treated with scrambled control PLK1S shRNA plasmids. CONCLUSIONS: The combination of shRNA-mediated gene silencing with effective in vivo gene delivery strategies appears to generate a long-lasting silencing signal.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases , RNA Nuclear Pequeno/administração & dosagem , RNA Nuclear Pequeno/farmacologia , Actinas/antagonistas & inibidores , Animais , Northern Blotting , Southern Blotting , Western Blotting , Proteínas de Ciclo Celular , DNA de Neoplasias/isolamento & purificação , Técnica Indireta de Fluorescência para Anticorpo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Camundongos , Camundongos Nus , Plasmídeos/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas , RNA Mensageiro/metabolismo , Transfecção , Transplante Heterólogo , Quinase 1 Polo-Like
18.
Proc Natl Acad Sci U S A ; 100(14): 8264-9, 2003 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-12826613

RESUMO

Reducing or eliminating expression of a given gene is likely to require multiple methods to ensure coverage of all of the genes in a given mammalian cell. We and others [Furth, P. A., Choe, W. T., Rex, J. H., Byrne, J. C., and Baker, C. C. (1994) Mol. Cell. Biol. 14, 5278-5289] have previously shown that U1 small nuclear (sn) RNA, both natural or with 5' end mutations, can specifically inhibit reporter gene expression in mammalian cells. This inhibition occurs when the U1 snRNA 5' end base pairs near the polyadenylation signal of the reporter gene's pre-mRNA. This base pairing inhibits poly(A) tail addition, a key, nearly universal step in mRNA biosynthesis, resulting in degradation of the mRNA. Here we demonstrate that expression of endogenous mammalian genes can be efficiently inhibited by transiently or stably expressed 5' end-mutated U1 snRNA. Also, we determine the inhibitory mechanism and establish a set of rules to use this technique and to improve the efficiency of inhibition. Two U1 snRNAs base paired to a single pre-mRNA act synergistically, resulting in up to 700-fold inhibition of the expression of specific reporter genes and 25-fold inhibition of endogenous genes. Surprisingly, distance from the U1 snRNA binding site to the poly(A) signal is not critical for inhibition, instead the U1 snRNA must be targeted to the terminal exon of the pre-mRNA. This could reflect a disruption by the 5' end-mutated U1 snRNA of the definition of the terminal exon as described by the exon definition model.


Assuntos
Precursores de RNA/antagonistas & inibidores , RNA Nuclear Pequeno/farmacologia , Regiões 3' não Traduzidas/genética , Regiões 5' não Traduzidas/genética , Pareamento de Bases , Sítios de Ligação , Sinergismo Farmacológico , Elementos Facilitadores Genéticos , Fibroblastos/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Células HeLa , Humanos , Íntrons/genética , Luciferases/genética , Conformação de Ácido Nucleico , Hibridização de Ácido Nucleico , Poli A/metabolismo , Regiões Promotoras Genéticas , Precursores de RNA/genética , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Splicing de RNA , RNA Nuclear Pequeno/genética , Vírus 40 dos Símios/genética
19.
World J Gastroenterol ; 9(3): 572-7, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12632521

RESUMO

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.


Assuntos
RNA Catalítico/farmacologia , RNA Nuclear Pequeno/farmacologia , RNA/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética , Animais , Linhagem Celular Transformada , Clonagem Molecular , Ratos , Fator de Crescimento Transformador beta1
20.
Blood ; 101(1): 104-11, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12393543

RESUMO

Mutations at nucleotides 654, 705, or 745 in intron 2 of the human beta-globin gene activate aberrant 3' and 5' splice sites within the intron and prevent correct splicing of beta-globin pre-mRNA, resulting in inhibition of beta-globin synthesis and in consequence beta-thalassemia. Transfection of HeLa cells expressing the 3 thalassemic mutants with modified U7 snRNA (U7.623), containing a sequence antisense to a region between the aberrant splice sites, reduced the incorrect splicing of pre-mRNA and led to increased levels of the correctly spliced beta-globin mRNA and protein. A lentiviral vector carrying the U7.623 gene was effective in restoration of correct splicing in the model cell lines for at least 6 months. Importantly, the therapeutic value of this system was demonstrated in hematopoietic stem cells and erythroid progenitor cells from a patient with IVS2-745/IVS2-1 thalassemia. Twelve days after transduction of the patient cells with the U7.623 lentiviral vector, the levels of correctly spliced beta-globin mRNA and hemoglobin A were approximately 25-fold over background. These results should be regarded as a proof of principle for lentiviral vector-based gene therapy for beta-thalassemia.


Assuntos
Células Precursoras Eritroides/metabolismo , Hemoglobina A/metabolismo , RNA Antissenso/farmacologia , RNA Nuclear Pequeno/farmacologia , Talassemia/terapia , Terapia Genética , Vetores Genéticos , Globinas/genética , Células HeLa , Hemoglobina A/genética , Humanos , Lentivirus/genética , Mutação , Splicing de RNA/efeitos dos fármacos , RNA Antissenso/administração & dosagem , RNA Antissenso/uso terapêutico , RNA Nuclear Pequeno/administração & dosagem , RNA Nuclear Pequeno/uso terapêutico , Talassemia/sangue , Talassemia/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...