Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
RNA ; 26(11): 1603-1620, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32675111

RESUMEN

Cellular quiescence and cell cycle reentry regulate vital biological processes such as cellular development and tissue homeostasis and are controlled by precise regulation of gene expression. The roles of long noncoding RNAs (lncRNAs) during these processes remain to be elucidated. By performing genome-wide transcriptome analyses, we identify differential expression of several hundreds of lncRNAs, including a significant number of the less-characterized class of microRNA-host-gene (MIRHG) lncRNAs or lnc-MIRHGs, during cellular quiescence and cell cycle reentry in human diploid fibroblasts. We observe that MIR222HG lncRNA displays serum-stimulated RNA processing due to enhanced splicing of the host nascent pri-MIR222HG transcript. The pre-mRNA splicing factor SRSF1 negatively regulates the microprocessor-catalyzed cleavage of pri-miR-222, thereby increasing the cellular pool of the mature MIR222HG Association of SRSF1 to pri-MIR222HG, including to a mini-exon, which partially overlaps with the primary miR-222 precursor, promotes serum-stimulated splicing over microRNA processing of MIR222HG Further, we observe that the increased levels of spliced MIR222HG in serum-stimulated cells promote the cell cycle reentry post quiescence in a microRNA-independent manner. MIR222HG interacts with DNM3OS, another lncRNA whose expression is elevated upon serum-stimulation, and promotes cell cycle reentry. The double-stranded RNA binding protein ILF3/2 complex facilitates MIR222HG:DNM3OS RNP complex assembly, thereby promoting DNM3OS RNA stability. Our study identifies a novel mechanism whereby competition between the splicing and microprocessor machinery modulates the serum-induced RNA processing of MIR222HG, which dictates cell cycle reentry.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Pulmón/citología , ARN Largo no Codificante/genética , Factores de Empalme Serina-Arginina/metabolismo , Suero/química , Ciclo Celular , Línea Celular , Fibroblastos/química , Fibroblastos/citología , Células HEK293 , Humanos , Pulmón/química , Proteína del Factor Nuclear 45/metabolismo , Proteínas del Factor Nuclear 90/metabolismo , Procesamiento Postranscripcional del ARN , Empalme del ARN , Análisis de Secuencia de ARN , Imagen Individual de Molécula , Regulación hacia Arriba , Secuenciación del Exoma
3.
Nucleic Acid Ther ; 30(4): 215-228, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32125928

RESUMEN

Reduction of matched and nearly complementary unintended transcripts was evaluated for 96 antisense oligonucleotides (ASOs) and 832 nearly matched unintended transcripts. The ASOs were 16-20 nucleotide "gapmers" with a gap of 8-10 DNA residues and 2'-O-methoxy-ethyl or constrained-ethyl substitutions in the wings. Most unintended transcripts were not reduced or were reduced with a potency more than 10-fold weaker than the intended transcript. For the unintended transcripts that were reduced, a strong correlation between relative potency of the intended versus the unintended transcript with predicted free energy of hybridization was observed. These results suggest ASO selectivity should be evaluated by testing for reduction of the unintended transcripts predicted to bind most stably to the ASO.


Asunto(s)
ADN/genética , Oligonucleótidos Antisentido/genética , Termodinámica , Transcriptoma/genética , Animales , Simulación por Computador , ADN/química , ADN/uso terapéutico , Humanos , Hibridación de Ácido Nucleico/métodos , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/uso terapéutico
4.
Haematologica ; 106(5): 1433-1442, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32439726

RESUMEN

ß-thalassemia is a disorder caused by altered hemoglobin protein synthesis and affects individuals worldwide. Severe forms of the disease, left untreated, can result in death before the age of 3 years (1). The standard of care consists of chronic and costly palliative treatment by blood transfusion combined with iron chelation. This dual approach suppresses anemia and reduces iron-related toxicities in patients. Allogeneic bone marrow transplant is an option, but limited by the availability of a highly compatible HSC donor. While gene therapy is been explored in several trials, its use is highly limited to developed regions with centers of excellence and well-established healthcare systems (2). Hence, there remains a tremendous unmet medical need to develop alternative treatment strategies for ß-thalassemia (3). Occurrence of aberrant splicing is one of the processes that affects ß-globin synthesis in ß-thalassemia. The (C>G) IVS-2-745 is a splicing mutation within intron 2 of the ß-globin gene. It leads to an aberrantly spliced mRNA that incorporates an intron fragment. This results in an in-frame premature termination codon that inhibits ß-globin production. Here, we propose the use of uniform 2'-O-methoxyethyl (2'-MOE) splice switching oligos (SSOs) to reverse this aberrant splicing in the pre-mRNA. With these lead SSOs we show aberrant to wild type splice switching. This switching leads to an increase of adult hemoglobin (HbA) up to 80% in erythroid cells from patients with the IVS-2-745 mutation. Furthermore, we demonstrate a restoration of the balance between ß-like- and α-globin chains, and up to an 87% reduction in toxic α-heme aggregates. While examining the potential benefit of 2'-MOE-SSOs in a mixed sickle-thalassemic phenotypic setting, we found reduced HbS synthesis and sickle cell formation due to HbA induction. In summary, 2'-MOE-SSOs are a promising therapy for forms of ß-thalassemia caused by mutations leading to aberrant splicing.

5.
PLoS Genet ; 14(11): e1007802, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30496290

RESUMEN

The human genome encodes thousands of long noncoding RNA (lncRNA) genes; the function of majority of them is poorly understood. Aberrant expression of a significant number of lncRNAs is observed in various diseases, including cancer. To gain insights into the role of lncRNAs in breast cancer progression, we performed genome-wide transcriptome analyses in an isogenic, triple negative breast cancer (TNBC/basal-like) progression cell lines using a 3D cell culture model. We identified significantly altered expression of 1853 lncRNAs, including ~500 natural antisense transcript (NATs) lncRNAs. A significant number of breast cancer-deregulated NATs displayed co-regulated expression with oncogenic and tumor suppressor protein-coding genes in cis. Further studies on one such NAT, PDCD4-AS1 lncRNA reveal that it positively regulates the expression and activity of the tumor suppressor PDCD4 in mammary epithelial cells. Both PDCD4-AS1 and PDCD4 show reduced expression in TNBC cell lines and in patients, and depletion of PDCD4-AS1 compromised the cellular levels and activity of PDCD4. Further, tumorigenic properties of PDCD4-AS1-depleted TNBC cells were rescued by exogenous expression of PDCD4, implying that PDCD4-AS1 acts upstream of PDCD4. Mechanistically, PDCD4-AS1 stabilizes PDCD4 RNA by forming RNA duplex and controls the interaction between PDCD4 RNA and RNA decay promoting factors such as HuR. Our studies demonstrate crucial roles played by NAT lncRNAs in regulating post-transcriptional gene expression of key oncogenic or tumor suppressor genes, thereby contributing to TNBC progression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Estabilidad del ARN , ARN sin Sentido/genética , ARN Largo no Codificante/genética , ARN Neoplásico/genética , Proteínas de Unión al ARN/genética , Neoplasias de la Mama Triple Negativas/genética , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Unión Proteica , ARN sin Sentido/metabolismo , ARN Largo no Codificante/metabolismo , ARN Neoplásico/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
6.
Nucleic Acids Res ; 46(19): 10405-10416, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30102375

RESUMEN

Long non-coding RNAs (lncRNAs) regulate vital biological processes, including cell proliferation, differentiation and development. A subclass of lncRNAs is synthesized from microRNA (miRNA) host genes (MIRHGs) due to pre-miRNA processing, and are categorized as miRNA-host gene lncRNAs (lnc-miRHGs). Presently, the cellular function of most lnc-miRHGs is not well understood. We demonstrate a miRNA-independent role for a nuclear-enriched lnc-miRHG in cell cycle progression. MIR100HG produces spliced and stable lncRNAs that display elevated levels during the G1 phase of the cell cycle. Depletion of MIR100HG-encoded lncRNAs in human cells results in aberrant cell cycle progression without altering the levels of miRNA encoded within MIR100HG. Notably, MIR100HG interacts with HuR/ELAVL1 as well as with several HuR-target mRNAs. Further, MIR100HG-depleted cells show reduced interaction between HuR and three of its target mRNAs, indicating that MIR100HG facilitates interaction between HuR and target mRNAs. Our studies have unearthed novel roles played by a MIRHG-encoded lncRNA in regulating RNA binding protein activity, thereby underscoring the importance of determining the function of several hundreds of lnc-miRHGs that are present in human genome.


Asunto(s)
Ciclo Celular/genética , Proteína 1 Similar a ELAV/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Diferenciación Celular/genética , División Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , ARN Mensajero/genética , Proteínas de Unión al ARN/genética
7.
Nat Genet ; 50(6): 814-824, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29808028

RESUMEN

The androgen receptor (AR) plays a critical role in the development of the normal prostate as well as prostate cancer. Using an integrative transcriptomic analysis of prostate cancer cell lines and tissues, we identified ARLNC1 (AR-regulated long noncoding RNA 1) as an important long noncoding RNA that is strongly associated with AR signaling in prostate cancer progression. Not only was ARLNC1 induced by the AR protein, but ARLNC1 stabilized the AR transcript via RNA-RNA interaction. ARLNC1 knockdown suppressed AR expression, global AR signaling and prostate cancer growth in vitro and in vivo. Taken together, these data support a role for ARLNC1 in maintaining a positive feedback loop that potentiates AR signaling during prostate cancer progression and identify ARLNC1 as a novel therapeutic target.


Asunto(s)
Neoplasias de la Próstata/genética , ARN Largo no Codificante/genética , Receptores Androgénicos/genética , Andrógenos/genética , Andrógenos/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Próstata/fisiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Largo no Codificante/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal
8.
Cell ; 171(7): 1559-1572.e20, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29245011

RESUMEN

Large-scale transcriptome sequencing efforts have vastly expanded the catalog of long non-coding RNAs (lncRNAs) with varying evolutionary conservation, lineage expression, and cancer specificity. Here, we functionally characterize a novel ultraconserved lncRNA, THOR (ENSG00000226856), which exhibits expression exclusively in testis and a broad range of human cancers. THOR knockdown and overexpression in multiple cell lines and animal models alters cell or tumor growth supporting an oncogenic role. We discovered a conserved interaction of THOR with IGF2BP1 and show that THOR contributes to the mRNA stabilization activities of IGF2BP1. Notably, transgenic THOR knockout produced fertilization defects in zebrafish and also conferred a resistance to melanoma onset. Likewise, ectopic expression of human THOR in zebrafish accelerated the onset of melanoma. THOR represents a novel class of functionally important cancer/testis lncRNAs whose structure and function have undergone positive evolutionary selection.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma/metabolismo , ARN Largo no Codificante/metabolismo , Pez Cebra , Animales , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Proteínas de Unión al ARN/metabolismo , Testículo/metabolismo
9.
J Cell Sci ; 130(24): 4180-4192, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29133588

RESUMEN

Nuclear speckles are self-assembled organelles composed of RNAs and proteins. They are proposed to act as structural domains that control distinct steps in gene expression, including transcription, splicing and mRNA export. Earlier studies identified differential localization of a few components within the speckles. It was speculated that the spatial organization of speckle components might contribute directly to the order of operations that coordinate distinct processes. Here, by performing multi-color structured illumination microscopy, we characterized the multilayer organization of speckles at a higher resolution. We found that SON and SC35 (also known as SRSF2) localize to the central region of the speckle, whereas MALAT1 and small nuclear (sn)RNAs are enriched at the speckle periphery. Coarse-grained simulations indicate that the non-random organization arises due to the interplay between favorable sequence-encoded intermolecular interactions of speckle-resident proteins and RNAs. Finally, we observe positive correlation between the total amount of RNA present within a speckle and the speckle size. These results imply that speckle size may be regulated to accommodate RNA accumulation and processing. Accumulation of RNA from various actively transcribed speckle-associated genes could contribute to the observed speckle size variations within a single cell.


Asunto(s)
Núcleo Celular/genética , Proteínas de Unión al ADN/genética , Antígenos de Histocompatibilidad Menor/genética , Orgánulos/genética , ARN Largo no Codificante/genética , Factores de Empalme Serina-Arginina/genética , Núcleo Celular/ultraestructura , Regulación de la Expresión Génica , Células HeLa , Humanos , Orgánulos/ultraestructura , Proteínas/genética , ARN/genética , ARN Nucleolar Pequeño/genética
10.
Hum Mol Genet ; 25(23): 5178-5187, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28007899

RESUMEN

ISCU myopathy is an inherited disease that primarily affects individuals of northern Swedish descent who share a single point mutation in the fourth intron of the ISCU gene. The current study shows correction of specific phenotypes associated with disease following treatment with an antisense oligonucleotide (ASO) targeted to the site of the mutation. We have shown that ASO treatment diminished aberrant splicing and increased ISCU protein levels in both patient fibroblasts and patient myotubes in a concentration dependent fashion. Upon ASO treatment, levels of SDHB in patient myotubular cell lines increased to levels observed in control myotubular cell lines. Additionally, we have shown that both patient fibroblast and myotubular cell lines displayed an increase in complex II activity with a concomitant decrease in succinate levels in patient myotubular cell lines after ASO treatment. Mitochondrial and cytosolic aconitase activities increased significantly following ASO treatment in patient myotubes. The current study suggests that ASO treatment may serve as a viable approach to correcting ISCU myopathy in patients.


Asunto(s)
Acidosis Láctica/congénito , Proteínas Hierro-Azufre/genética , Enfermedades Musculares/congénito , Oligonucleótidos Antisentido/genética , Succinato Deshidrogenasa/genética , Acidosis Láctica/genética , Acidosis Láctica/patología , Acidosis Láctica/terapia , Femenino , Humanos , Intrones/genética , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/terapia , Oligonucleótidos Antisentido/uso terapéutico , Fenotipo , Mutación Puntual , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Succinato Deshidrogenasa/biosíntesis
11.
Cell Rep ; 17(1): 261-274, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681436

RESUMEN

Long non-coding RNAs (lncRNAs) represent the largest and most diverse class of non-coding RNAs, comprising almost 16,000 currently annotated transcripts in human and 10,000 in mouse. Here, we investigated the role of lncRNAs in mammary tumors by performing RNA-seq on tumor sections and organoids derived from MMTV-PyMT and MMTV-Neu-NDL mice. We identified several hundred lncRNAs that were overexpressed compared to normal mammary epithelium. Among these potentially oncogenic lncRNAs we prioritized a subset as Mammary Tumor Associated RNAs (MaTARs) and determined their human counterparts, hMaTARs. To functionally validate the role of MaTARs, we performed antisense knockdown and observed reduced cell proliferation, invasion, and/or organoid branching in a cancer-specific context. Assessing the expression of hMaTARs in human breast tumors revealed that 19 hMaTARs are significantly upregulated and many of these correlate with breast cancer subtype and/or hormone receptor status, indicating potential clinical relevance.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Mamarias Animales/terapia , Oligorribonucleótidos Antisentido/genética , ARN Largo no Codificante/genética , ARN Neoplásico/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Femenino , Humanos , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Transgénicos , Oligorribonucleótidos Antisentido/metabolismo , Oligorribonucleótidos Antisentido/uso terapéutico , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/metabolismo , ARN Neoplásico/antagonistas & inhibidores , ARN Neoplásico/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Transcriptoma
12.
Neoplasia ; 18(8): 489-99, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27566105

RESUMEN

Rapid advances in the discovery of long noncoding RNAs (lncRNAs) have identified lineage- and cancer-specific biomarkers that may be relevant in the clinical management of prostate cancer (PCa). Here we assembled and analyzed a large RNA-seq dataset, from 585 patient samples, including benign prostate tissue and both localized and metastatic PCa to discover and validate differentially expressed genes associated with disease aggressiveness. We performed Sample Set Enrichment Analysis (SSEA) and identified genes associated with low versus high Gleason score in the RNA-seq database. Comparing Gleason 6 versus 9+ PCa samples, we identified 99 differentially expressed genes with variable association to Gleason grade as well as robust expression in prostate cancer. The top-ranked novel lncRNA PCAT14, exhibits both cancer and lineage specificity. On multivariate analysis, low PCAT14 expression independently predicts for BPFS (P=.00126), PSS (P=.0385), and MFS (P=.000609), with trends for OS as well (P=.056). An RNA in-situ hybridization (ISH) assay for PCAT14 distinguished benign vs malignant cases, as well as high vs low Gleason disease. PCAT14 is transcriptionally regulated by AR, and endogenous PCAT14 overexpression suppresses cell invasion. Thus, Using RNA-sequencing data we identify PCAT14, a novel prostate cancer and lineage-specific lncRNA. PCAT14 is highly expressed in low grade disease and loss of PCAT14 predicts for disease aggressiveness and recurrence.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/mortalidad , ARN Largo no Codificante/genética , Línea Celular Tumoral , Análisis por Conglomerados , Progresión de la Enfermedad , Estudios de Seguimiento , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Hibridación in Situ , Estimación de Kaplan-Meier , Masculino , Clasificación del Tumor , Recurrencia Local de Neoplasia , Pronóstico , Neoplasias de la Próstata/patología , Transporte de ARN , ARN Largo no Codificante/metabolismo , Reproducibilidad de los Resultados
13.
Oncotarget ; 7(26): 40418-40436, 2016 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-27250026

RESUMEN

MALAT1 (metastasis associated lung adenocarcinoma transcript1) is a conserved long non-coding RNA, known to regulate gene expression by modulating transcription and post-transcriptional pre-mRNA processing of a large number of genes. MALAT1 expression is deregulated in various tumors, including breast cancer. However, the significance of such abnormal expression is yet to be fully understood. In this study, we demonstrate that regulation of aggressive breast cancer cell traits by MALAT1 is not predicted solely based on an elevated expression level but is context specific. By performing loss- and gain-of-function studies, both under in vitro and in vivo conditions, we demonstrate that MALAT1 facilitates cell proliferation, tumor progression and metastasis of triple-negative breast cancer (TNBC) cells despite having a comparatively lower expression level than ER or HER2-positive breast cancer cells. Furthermore, MALAT1 regulates the expression of several cancer metastasis-related genes, but displays molecular subtype specific correlations with such genes. Assessment of the prognostic significance of MALAT1 in human breast cancer (n=1992) revealed elevated MALAT1 expression was associated with decreased disease-specific survival in ER negative, lymph node negative patients of the HER2 and TNBC molecular subtypes. Multivariable analysis confirmed MALAT1 to have independent prognostic significance in the TNBC lymph node negative patient subset (HR=2.64, 95%CI 1.35- 5.16, p=0.005). We propose that the functional significance of MALAT1 as a metastasis driver and its potential use as a prognostic marker is most promising for those patients diagnosed with ER negative, lymph node negative breast cancer who might otherwise mistakenly be stratified to have low recurrence risk.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , ARN Largo no Codificante/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Anisomicina/química , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Análisis Multivariante , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Fenotipo , Pronóstico , Empalme del ARN , Riesgo
14.
Nucleic Acids Res ; 44(6): 2898-908, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26826711

RESUMEN

The RNase P-mediated endonucleolytic cleavage plays a crucial role in the 3' end processing and cellular accumulation of MALAT1, a nuclear-retained long noncoding RNA that promotes malignancy. The regulation of this cleavage event is largely undetermined. Here we characterize a broadly expressed natural antisense transcript at the MALAT1 locus, designated as TALAM1, that positively regulates MALAT1 levels by promoting the 3' end cleavage and maturation of MALAT1 RNA. TALAM1 RNA preferentially localizes at the site of transcription, and also interacts with MALAT1 RNA. Depletion of TALAM1 leads to defects in the 3' end cleavage reaction and compromises cellular accumulation of MALAT1. Conversely, overexpression of TALAM1 facilitates the cleavage reaction in trans Interestingly, TALAM1 is also positively regulated by MALAT1 at the level of both transcription and RNA stability. Together, our data demonstrate a novel feed-forward positive regulatory loop that is established to maintain the high cellular levels of MALAT1, and also unravel the existence of sense-antisense mediated regulatory mechanism for cellular lncRNAs that display RNase P-mediated 3' end processing.


Asunto(s)
Núcleo Celular/metabolismo , ARN sin Sentido/genética , ARN Largo no Codificante/antagonistas & inhibidores , Secuencia de Bases , Línea Celular Tumoral , Regulación de la Expresión Génica , Células HCT116 , Células HeLa , Humanos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , División del ARN , Estabilidad del ARN , ARN sin Sentido/química , ARN sin Sentido/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal
15.
Nucleic Acids Res ; 44(5): 2093-109, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26553810

RESUMEN

High affinity antisense oligonucleotides (ASOs) containing bicylic modifications (BNA) such as locked nucleic acid (LNA) designed to induce target RNA cleavage have been shown to have enhanced potency along with a higher propensity to cause hepatotoxicity. In order to understand the mechanism of this hepatotoxicity, transcriptional profiles were collected from the livers of mice treated with a panel of highly efficacious hepatotoxic or non-hepatotoxic LNA ASOs. We observed highly selective transcript knockdown in mice treated with non-hepatotoxic LNA ASOs, while the levels of many unintended transcripts were reduced in mice treated with hepatotoxic LNA ASOs. This transcriptional signature was concurrent with on-target RNA reduction and preceded transaminitis. Remarkably, the mRNA transcripts commonly reduced by toxic LNA ASOs were generally not strongly associated with any particular biological process, cellular component or functional group. However, they tended to have much longer pre-mRNA transcripts. We also demonstrate that the off-target RNA knockdown and hepatotoxicity is attenuated by RNase H1 knockdown, and that this effect can be generalized to high affinity modifications beyond LNA. This suggests that for a certain set of ASOs containing high affinity modifications such as LNA, hepatotoxicity can occur as a result of unintended off-target RNase H1 dependent RNA degradation.


Asunto(s)
Hígado/efectos de los fármacos , Oligonucleótidos Antisentido/toxicidad , Oligonucleótidos/toxicidad , ARN Mensajero/genética , Ribonucleasa H/genética , Alanina Transaminasa/sangre , Alanina Transaminasa/genética , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Análisis por Micromatrices , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Precursores del ARN/antagonistas & inhibidores , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ribonucleasa H/antagonistas & inhibidores , Ribonucleasa H/metabolismo , Transcriptoma/efectos de los fármacos
16.
J Pharmacol Exp Ther ; 355(2): 329-40, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26330536

RESUMEN

Myotonic dystrophy type 1 (DM1) is the most common form of muscular dystrophy in adults. DM1 is caused by an expanded CTG repeat in the 3'-untranslated region of DMPK, the gene encoding dystrophia myotonica protein kinase (DMPK). Antisense oligonucleotides (ASOs) containing 2',4'-constrained ethyl-modified (cEt) residues exhibit a significantly increased RNA binding affinity and in vivo potency relative to those modified with other 2'-chemistries, which we speculated could translate to enhanced activity in extrahepatic tissues, such as muscle. Here, we describe the design and characterization of a cEt gapmer DMPK ASO (ISIS 486178), with potent activity in vitro and in vivo against mouse, monkey, and human DMPK. Systemic delivery of unformulated ISIS 486718 to wild-type mice decreased DMPK mRNA levels by up to 90% in liver and skeletal muscle. Similarly, treatment of either human DMPK transgenic mice or cynomolgus monkeys with ISIS 486178 led to up to 70% inhibition of DMPK in multiple skeletal muscles and ∼50% in cardiac muscle in both species. Importantly, inhibition of DMPK was well tolerated and was not associated with any skeletal muscle or cardiac toxicity. Also interesting was the demonstration that the inhibition of DMPK mRNA levels in muscle was maintained for up to 16 and 13 weeks post-treatment in mice and monkeys, respectively. These results demonstrate that cEt-modified ASOs show potent activity in skeletal muscle, and that this attractive therapeutic approach warrants further clinical investigation to inhibit the gain-of-function toxic RNA underlying the pathogenesis of DM1.


Asunto(s)
Distrofia Miotónica/tratamiento farmacológico , Proteína Quinasa de Distrofia Miotónica/metabolismo , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos/farmacología , Animales , Línea Celular , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/enzimología , Proteína Quinasa de Distrofia Miotónica/antagonistas & inhibidores , Proteína Quinasa de Distrofia Miotónica/genética , Oligonucleótidos/química , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
17.
PLoS One ; 10(7): e0132798, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26207833

RESUMEN

Liver regeneration after partial hepatectomy (PHx) is a complex and well-orchestrated biological process in which synchronized cell proliferation is induced in response to the loss of liver mass. To define long noncoding RNAs (lncRNAs) that participate in the regulation of liver regeneration, we performed microarray analysis and identified more than 400 lncRNAs exhibiting significantly altered expression. Of these, one lncRNA, LncPHx2 (Long noncoding RNA induced by PHx 2), was highly upregulated during liver regeneration. Depletion of LncPHx2 during liver regeneration using antisense oligonucleotides led to a transient increase in hepatocyte proliferation and more rapid liver regeneration. Gene expression analysis showed that LncPHx2 depletion resulted in upregulation of mRNAs encoding proteins known to promote cell proliferation, including MCM components, DNA polymerases, histone proteins, and transcription factors. LncPHx2 interacts with the mRNAs of MCM components, making it a candidate to regulate the expression of MCMs and other genes post-transcriptionally. Collectively, our data demonstrate that LncPHx2 is a key lncRNA that participates in a negative feedback loop modulating hepatocyte proliferation through RNA-RNA interactions.


Asunto(s)
Proliferación Celular/genética , Hepatectomía , Hepatocitos/fisiología , Regeneración Hepática/genética , ARN Largo no Codificante/fisiología , Animales , Células Cultivadas , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , ARN Largo no Codificante/genética , Regulación hacia Arriba/genética
18.
Genome Res ; 25(9): 1336-46, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26048247

RESUMEN

Long noncoding (lnc)RNAs have recently emerged as key regulators of gene expression. Here, we performed high-depth poly(A)(+) RNA sequencing across multiple clonal populations of mouse embryonic stem cells (ESCs) and neural progenitor cells (NPCs) to comprehensively identify differentially regulated lncRNAs. We establish a biologically robust profile of lncRNA expression in these two cell types and further confirm that the majority of these lncRNAs are enriched in the nucleus. Applying weighted gene coexpression network analysis, we define a group of lncRNAs that are tightly associated with the pluripotent state of ESCs. Among these, we show that acute depletion of Platr14 using antisense oligonucleotides impacts the differentiation- and development-associated gene expression program of ESCs. Furthermore, we demonstrate that Firre, a lncRNA highly enriched in the nucleoplasm and previously reported to mediate chromosomal contacts in ESCs, controls a network of genes related to RNA processing. Together, we provide a comprehensive, up-to-date, and high resolution compilation of lncRNA expression in ESCs and NPCs and show that nuclear lncRNAs are tightly integrated into the regulation of ESC gene expression.


Asunto(s)
Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , ARN Largo no Codificante/genética , Transcriptoma , Animales , Diferenciación Celular/genética , Núcleo Celular , Análisis por Conglomerados , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo
19.
Methods Mol Biol ; 1262: 321-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25555591

RESUMEN

Long noncoding RNAs (lncRNAs) have recently emerged as important players in diverse cellular processes. Among them, a large fraction of lncRNAs are localized within cell nucleus. And several of these nuclear-retained lncRNAs have been found to regulate key nuclear processes, which brings up the requirement of effective genetic tools to explore the functions of this "dark matter" inside the nucleus. While siRNAs and shRNAs are widely used tools in loss-of-function studies, their general efficiency in depleting nuclear-retained lncRNAs is limited, due to the fact that the RNAi machinery is located mainly in the cytoplasm of mammalian cells. Here, we describe the usage of chemically modified chimeric DNA antisense oligonucleotides (ASO) in effective knockdown of nuclear-retained lncRNAs, with a focus on the detailed workflow from the design and synthesis of ASOs, to in vitro and in vivo delivery methods.


Asunto(s)
Oligonucleótidos Antisentido/química , ARN Largo no Codificante/genética , ARN Mensajero Almacenado/genética , Animales , Línea Celular , Núcleo Celular/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Células Hep G2 , Humanos , Oligonucleótidos Antisentido/genética
20.
PLoS One ; 9(10): e110615, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25334092

RESUMEN

A new strategy for identifying potent RNase H-dependent antisense oligonucleotides (ASOs) is presented. Our analysis of the human transcriptome revealed that a significant proportion of genes contain unique repeated sequences of 16 or more nucleotides in length. Activities of ASOs targeting these repeated sites in several representative genes were compared to those of ASOs targeting unique single sites in the same transcript. Antisense activity at repeated sites was also evaluated in a highly controlled minigene system. Targeting both native and minigene repeat sites resulted in significant increases in potency as compared to targeting of non-repeated sites. The increased potency at these sites is a result of increased frequency of ASO/RNA interactions which, in turn, increases the probability of a productive interaction between the ASO/RNA heteroduplex and human RNase H1 in the cell. These results suggest a new, highly efficient strategy for rapid identification of highly potent ASOs.


Asunto(s)
Oligonucleótidos Antisentido/genética , Secuencias Repetitivas de Ácidos Nucleicos , Sitios de Unión , Línea Celular , Expresión Génica , Genes Reporteros , Humanos , Oligonucleótidos Antisentido/metabolismo , Empalme del ARN , Receptores de Glucagón/genética , Ribonucleasa H/metabolismo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...