Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2537: 335-350, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35895273

RESUMEN

We describe methods to follow the fate of oligonucleotides after their injection into experimental animals. The quantitation in various tissues, blood or bone marrow cells is possible by chemical ligation PCR. This method works independently of chemical modifications of the oligonucleotide and/or its conjugations to lipid or peptide moieties. Moreover, metabolization intermediates can be detected by mass spectrometry. Together with a readout assay for the biochemical or physiological effects, which will differ, depending on the particular purpose of the oligonucleotide, these methods allow for a comprehensive understanding of oligonucleotide behavior in a living organism.


Asunto(s)
Oligonucleótidos , Péptidos , Animales , Espectrometría de Masas/métodos , Oligonucleótidos/química , Péptidos/química , Distribución Tisular
2.
Cell Chem Biol ; 28(8): 1221-1234.e6, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-33756123

RESUMEN

Erythropoietic protoporphyria (EPP) is a rare disease in which patients experience severe light sensitivity. It is caused by a deficiency of ferrochelatase (FECH), the last enzyme in heme biosynthesis (HBS). The lack of FECH causes accumulation of its photoreactive substrate protoporphyrin IX (PPIX) in patients' erythrocytes. Here, we explored an approach for the treatment of EPP by decreasing PPIX synthesis using small-molecule inhibitors directed to factors in the HBS pathway. We generated a FECH-knockout clone from K562 erythroleukemia cells, which accumulates PPIX and undergoes oxidative stress upon light exposure. We used these matched cell lines to screen a set of publicly available inhibitors of factors in the HBS pathway. Inhibitors of the glycine transporters GlyT1 and GlyT2 lowered levels of PPIX and markers of oxidative stress selectively in K56211B4 cells, and in primary erythroid cultures from an EPP patient. Our findings open the door to investigation of glycine transport inhibitors for HBS disorders.


Asunto(s)
Proteínas de Transporte de Glicina en la Membrana Plasmática/antagonistas & inhibidores , Protoporfiria Eritropoyética/tratamiento farmacológico , Protoporfirinas/farmacología , Células Cultivadas , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Humanos , Células K562 , Estructura Molecular , Protoporfiria Eritropoyética/metabolismo
3.
Nucleic Acids Res ; 48(9): 4658-4671, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32313951

RESUMEN

Erythropoietic protoporphyria (EPP) is a rare genetic disease in which patients experience acute phototoxic reactions after sunlight exposure. It is caused by a deficiency in ferrochelatase (FECH) in the heme biosynthesis pathway. Most patients exhibit a loss-of-function mutation in trans to an allele bearing a SNP that favors aberrant splicing of transcripts. One viable strategy for EPP is to deploy splice-switching oligonucleotides (SSOs) to increase FECH synthesis, whereby an increase of a few percent would provide therapeutic benefit. However, successful application of SSOs in bone marrow cells is not described. Here, we show that SSOs comprising methoxyethyl-chemistry increase FECH levels in cells. We conjugated one SSO to three prototypical targeting groups and administered them to a mouse model of EPP in order to study their biodistribution, their metabolic stability and their FECH splice-switching ability. The SSOs exhibited distinct distribution profiles, with increased accumulation in liver, kidney, bone marrow and lung. However, they also underwent substantial metabolism, mainly at their linker groups. An SSO bearing a cholesteryl group increased levels of correctly spliced FECH transcript by 80% in the bone marrow. The results provide a promising approach to treat EPP and other disorders originating from splicing dysregulation in the bone marrow.


Asunto(s)
Ferroquelatasa/genética , Oligonucleótidos/administración & dosificación , Protoporfiria Eritropoyética/metabolismo , Empalme del ARN , Albúminas/metabolismo , Animales , Médula Ósea/metabolismo , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Ferroquelatasa/metabolismo , Humanos , Células K562 , Ratones , Oligonucleótidos/sangre , Oligonucleótidos/química , Oligonucleótidos/farmacocinética , Polimorfismo de Nucleótido Simple , Protoporfiria Eritropoyética/genética , Protoporfiria Eritropoyética/terapia , Sitios de Empalme de ARN , Distribución Tisular
4.
Mol Genet Metab ; 128(3): 304-308, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31076252

RESUMEN

Deficiency in ferrochelatase (FECH), the last enzyme in the heme biosynthetic pathway, leads to an accumulation of protoporphyrin IX (PPIX) that causes a severely painful phototoxic reaction of the skin in patients with erythropoietic protoporphyria (EPP). Besides phototoxicity of the skin, EPP patients often present with symptoms of iron deficiency in form of a microcytic and hypochromic anemia with low serum iron and ferritin. In addition, elevated aminolevulinic acid synthase 2 (ALAS2) both at the mRNA and protein levels have been observed among EPP patients. ALAS is the first enzyme in the pathway and exists in two isoforms, whereby the isoform 2 (ALAS2) is expressed exclusively in erythropoiesis. The mRNA of ALAS2 contains an iron response element (IRE) at its 5'UTR. When iron is limited, iron response element binding protein 2 (IRP2) binds to the IRE of ALAS2 mRNA and suppresses its translation. In this study, we demonstrated that iron deprivation increased the amount of ALAS2 mRNA as well as the ratio of ALAS2 to FECH mRNAs in cultured erythroleukemic K562 cells. At the protein level, however, iron deprivation in the cell line caused reductions in both enzymes as shown by the Western blot analysis. A comparable increase in the ratio of ALAS2 to FECH mRNAs was also found in EPP patients indicating an imbalance in heme biosynthesis. As iron cannot be completely missing from an organism, we assume that in EPP patients, a certain amount of ALAS2 mRNA is translated despite a partial deficiency of FECH. The increase in ALAS2 enzyme contributes to the accumulation in PPIX in the patients. Targeted inhibition of ALAS2 could therefore be a treatment option for EPP.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , Hierro/metabolismo , Protoporfiria Eritropoyética/enzimología , Vías Biosintéticas , Ferroquelatasa/genética , Humanos , Hierro/sangre , Proteína 2 Reguladora de Hierro/metabolismo , Proteínas Reguladoras del Hierro/metabolismo , Células K562 , Protoporfiria Eritropoyética/terapia , Protoporfirinas/metabolismo
5.
BMC Mol Biol ; 19(1): 9, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-30053800

RESUMEN

BACKGROUND: Core canonical histones are required in the S phase of the cell cycle to pack newly synthetized DNA, therefore the expression of their genes is highly activated during DNA replication. In mammalian cells, this increment is achieved by both enhanced transcription and 3' end processing. In this paper, we described positive cofactor 4 (PC4) as a protein that contributes to the regulation of replication-dependent histone gene expression. RESULTS: We showed that PC4 influences RNA polymerase II recruitment to histone gene loci in a cell cycle-dependent manner. The most important effect was observed in S phase where PC4 knockdown leads to the elevated level of RNA polymerase II on histone genes, which corresponds to the increased total level of those gene transcripts. The opposite effect was caused by PC4 overexpression. Moreover, we found that PC4 has a negative effect on the unique 3' end processing of histone pre-mRNAs that can be based on the interaction of PC4 with U7 snRNP and CstF64. Interestingly, this effect does not depend on the cell cycle. CONCLUSIONS: We conclude that PC4 might repress RNA polymerase II recruitment and transcription of replication-dependent histone genes in order to maintain the very delicate balance between histone gene expression and DNA synthesis. It guards the cell from excess of histones in S phase. Moreover, PC4 might promote the interaction of cleavage and polyadenylation complex with histone pre-mRNAs, that might impede with the recruitment of histone cleavage complex. This in turn decreases the 3' end processing efficiency of histone gene transcripts.


Asunto(s)
Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Histonas/genética , ARN Polimerasa II/metabolismo , Factores de Transcripción/metabolismo , Ciclo Celular , Factor de Estimulación del Desdoblamiento/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Procesamiento de Término de ARN 3' , Ribonucleoproteína Nuclear Pequeña U7/metabolismo
6.
Dis Model Mech ; 10(3): 225-233, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28093505

RESUMEN

Erythropoietic protoporphyria (EPP) is caused by deficiency of ferrochelatase (FECH), which incorporates iron into protoporphyrin IX (PPIX) to form heme. Excitation of accumulated PPIX by light generates oxygen radicals that evoke excessive pain and, after longer light exposure, cause ulcerations in exposed skin areas of individuals with EPP. Moreover, ∼5% of the patients develop a liver dysfunction as a result of PPIX accumulation. Most patients (∼97%) have a severe FECH mutation (Mut) in trans to an intronic polymorphism (c.315-48C), which reduces ferrochelatase synthesis by stimulating the use of an aberrant 3' splice site 63 nt upstream of the normal site for exon 4. In contrast, with the predominant c.315-48T allele, the correct splice site is mostly used, and individuals with a T/Mut genotype do not develop EPP symptoms. Thus, the C allele is a potential target for therapeutic approaches that modify this splicing decision. To provide a model for pre-clinical studies of such approaches, we engineered a mouse containing a partly humanized Fech gene with the c.315-48C polymorphism. F1 hybrids obtained by crossing these mice with another inbred line carrying a severe Fech mutation (named m1Pas) show a very strong EPP phenotype that includes elevated PPIX in the blood, enlargement of liver and spleen, anemia, as well as strong pain reactions and skin lesions after a short period of light exposure. In addition to the expected use of the aberrant splice site, the mice also show a strong skipping of the partly humanized exon 3. This will limit the use of this model for certain applications and illustrates that engineering of a hybrid gene may have unforeseeable consequences on its splicing.


Asunto(s)
Ferroquelatasa/genética , Mutación/genética , Protoporfiria Eritropoyética/enzimología , Protoporfiria Eritropoyética/genética , Alelos , Empalme Alternativo/genética , Animales , Secuencia de Bases , Células Sanguíneas/patología , Cruzamiento , Modelos Animales de Enfermedad , Exones/genética , Genotipo , Recombinación Homóloga/genética , Humanos , Luz , Hígado/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamaño de los Órganos , Protoporfiria Eritropoyética/sangre , Protoporfiria Eritropoyética/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Piel/patología , Piel/efectos de la radiación
7.
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
9.
Curr Opin Cell Biol ; 40: 23-31, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26895140

RESUMEN

The histones which pack new DNA during the S phase of animal cells are made from mRNAs that are cleaved at their 3' end but not polyadenylated. Some of the factors used in this reaction are unique to it while others are shared with the polyadenylation process that generates all other mRNAs. Recent work has begun to shed light on how the cell manages the assignment of these common components to the two 3' processing systems, and how it achieves their cell cycle-regulation and recruitment to the histone pre-mRNA. Moreover, recent and older findings reveal multiple connections between the nuclear organization of histone genes, their transcription and 3' end processing as well as the control of cell proliferation.


Asunto(s)
Histonas/genética , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Animales , Ciclo Celular , Puntos de Control del Ciclo Celular , Núcleo Celular/metabolismo , Replicación del ADN , Expresión Génica , Humanos , Poliadenilación
10.
Mol Cell Neurosci ; 70: 30-41, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26621405

RESUMEN

Spinal muscular atrophy (SMA) is characterized by motoneuron loss and muscle weakness. However, the structural and functional deficits that lead to the impairment of the neuromuscular system remain poorly defined. By electron microscopy, we previously found that neuromuscular junctions (NMJs) and muscle fibres of the diaphragm are among the earliest affected structures in the severe mouse SMA model. Because of certain anatomical features, i.e. its thinness and its innervation from the cervical segments of the spinal cord, the diaphragm is particularly suitable to characterize both central and peripheral events. Here we show by immunohistochemistry that, at postnatal day 3, the cervical motoneurons of SMA mice receive less stimulatory synaptic inputs. Moreover, their mitochondria become less elongated which might represent an early stage of degeneration. The NMJs of the diaphragm of SMA mice show a loss of synaptic vesicles and active zones. Moreover, the partly innervated endplates lack S100 positive perisynaptic Schwann cells (PSCs). We also demonstrate the feasibility of comparing the proteomic composition between diaphragm regions enriched and poor in NMJs. By this approach we have identified two proteins that are significantly upregulated only in the NMJ-specific regions of SMA mice. These are apoptosis inducing factor 1 (AIFM1), a mitochondrial flavoprotein that initiates apoptosis in a caspase-independent pathway, and four and a half Lim domain protein 1 (FHL1), a regulator of skeletal muscle mass that has been implicated in several myopathies.


Asunto(s)
Diafragma/inervación , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Factor Inductor de la Apoptosis/metabolismo , Diafragma/metabolismo , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Ratones , Proteínas Musculares/metabolismo , Atrofia Muscular Espinal/fisiopatología , Unión Neuromuscular/metabolismo , Proteómica , Células de Schwann/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Vesículas Sinápticas/metabolismo
11.
Nucleic Acids Res ; 43(20): 9711-28, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26250115

RESUMEN

Replication-dependent histone genes are up-regulated during the G1/S phase transition to meet the requirement for histones to package the newly synthesized DNA. In mammalian cells, this increment is achieved by enhanced transcription and 3' end processing. The non-polyadenylated histone mRNA 3' ends are generated by a unique mechanism involving the U7 small ribonucleoprotein (U7 snRNP). By using affinity purification methods to enrich U7 snRNA, we identified FUS/TLS as a novel U7 snRNP interacting protein. Both U7 snRNA and histone transcripts can be precipitated by FUS antibodies predominantly in the S phase of the cell cycle. Moreover, FUS depletion leads to decreased levels of correctly processed histone mRNAs and increased levels of extended transcripts. Interestingly, FUS antibodies also co-immunoprecipitate histone transcriptional activator NPAT and transcriptional repressor hnRNP UL1 in different phases of the cell cycle. We further show that FUS binds to histone genes in S phase, promotes the recruitment of RNA polymerase II and is important for the activity of histone gene promoters. Thus, FUS may serve as a linking factor that positively regulates histone gene transcription and 3' end processing by interacting with the U7 snRNP and other factors involved in replication-dependent histone gene expression.


Asunto(s)
Replicación del ADN , Regulación de la Expresión Génica , Histonas/genética , Proteína FUS de Unión a ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U7/metabolismo , Transcripción Genética , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Células HEK293 , Células HeLa , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Histonas/biosíntesis , Humanos , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , ARN Nuclear Pequeño/metabolismo , Factores de Transcripción/metabolismo
12.
Ann Neurol ; 77(3): 399-414, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25516063

RESUMEN

OBJECTIVES: Spinal muscular atrophy (SMA) is caused by reduced levels of survival motor neuron (SMN) protein, which results in motoneuron loss. Therapeutic strategies to increase SMN levels including drug compounds, antisense oligonucleotides, and scAAV9 gene therapy have proved effective in mice. We wished to determine whether reduction of SMN in postnatal motoneurons resulted in SMA in a large animal model, whether SMA could be corrected after development of muscle weakness, and the response of clinically relevant biomarkers. METHODS: Using intrathecal delivery of scAAV9 expressing an shRNA targeting pig SMN1, SMN was knocked down in motoneurons postnatally to SMA levels. This resulted in an SMA phenotype representing the first large animal model of SMA. Restoration of SMN was performed at different time points with scAAV9 expressing human SMN (scAAV9-SMN), and electrophysiology measurements and pathology were performed. RESULTS: Knockdown of SMN in postnatal motoneurons results in overt proximal weakness, fibrillations on electromyography indicating active denervation, and reduced compound muscle action potential (CMAP) and motor unit number estimation (MUNE), as in human SMA. Neuropathology showed loss of motoneurons and motor axons. Presymptomatic delivery of scAAV9-SMN prevented SMA symptoms, indicating that all changes are SMN dependent. Delivery of scAAV9-SMN after symptom onset had a marked impact on phenotype, electrophysiological measures, and pathology. INTERPRETATION: High SMN levels are critical in postnatal motoneurons, and reduction of SMN results in an SMA phenotype that is SMN dependent. Importantly, clinically relevant biomarkers including CMAP and MUNE are responsive to SMN restoration, and abrogation of phenotype can be achieved even after symptom onset.


Asunto(s)
Modelos Animales de Enfermedad , Terapia Genética/métodos , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/terapia , Proteínas del Complejo SMN/metabolismo , Animales , Biomarcadores , Dependovirus/genética , Electromiografía , Vectores Genéticos/uso terapéutico , Humanos , Neuronas Motoras/patología , Atrofia Muscular Espinal/etiología , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Fenotipo , ARN Interferente Pequeño/uso terapéutico , Proteínas del Complejo SMN/genética , Porcinos
13.
Mol Cell Biol ; 34(23): 4272-84, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25266659

RESUMEN

The 3' end processing of animal replication-dependent histone mRNAs is activated during G1/S-phase transition. The processing site is recognized by stem-loop binding protein and the U7 snRNP, but cleavage additionally requires a heat-labile factor (HLF), composed of cleavage/polyadenylation specificity factor, symplekin, and cleavage stimulation factor 64 (CstF64). Although HLF has been shown to be cell cycle regulated, the mechanism of this regulation is unknown. Here we show that levels of CstF64 increase toward the S phase and its depletion affects histone RNA processing, S-phase progression, and cell proliferation. Moreover, analyses of the interactions between CstF64, symplekin, and the U7 snRNP-associated proteins FLASH and Lsm11 indicate that CstF64 is important for recruiting HLF to histone precursor mRNA (pre-mRNA)-resident proteins. Thus, CstF64 is central to the function of HLF and appears to be at least partly responsible for its cell cycle regulation. Additionally, we show that misprocessed histone transcripts generated upon CstF64 depletion mainly accumulate in the nucleus, where they are targets of the exosome machinery, while a small cytoplasmic fraction is partly associated with polysomes.


Asunto(s)
Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Histonas/genética , Proteínas Nucleares/genética , Procesamiento de Término de ARN 3'/genética , Proteínas de Unión al ARN/fisiología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Proliferación Celular , Factor de Estimulación del Desdoblamiento , Células HEK293 , Células HeLa , Humanos , Precursores del ARN/genética , Proteínas de Unión al ARN/genética , Ribonucleoproteína Nuclear Pequeña U7/genética , Fase S/genética
14.
Neuropathol Appl Neurobiol ; 40(4): 416-34, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23718187

RESUMEN

AIMS: As 4-day-old mice of the severe spinal muscular atrophy (SMA) model (dying at 5-8 days) display pronounced neuromuscular changes in the diaphragm but not the soleus muscle, we wanted to gain more insight into the relationship between muscle development and the emergence of pathological changes and additionally to analyse intercostal muscles which are affected in human SMA. METHODS: Structures of muscle fibres and neuromuscular junctions (NMJs) of the diaphragm, intercostal and calf muscles of prenatal (E21) and postnatal (P0 and P4) healthy and SMA mice were analysed by light and transmission electron microscopy. NMJ innervation was studied by whole mount immunofluorescence in diaphragms of P4 mice. RESULTS: During this period, the investigated muscles still show a significant neck-to-tail developmental gradient. The diaphragm and calf muscles are most and least advanced, respectively, with respect to muscle fibre fusion and differentiation. The number and depth of subsynaptic folds increases, and perisynaptic Schwann cells (PSCs) acquire a basal lamina on their outer surface. Subsynaptic folds are connected to an extensive network of tubules and beaded caveolae, reminiscent of the T system in adult muscle. Interestingly, intercostal muscles from P4 SMA mice show weaker pathological involvement (that is, vacuolization of PSCs and perineurial cells) than those previously described by us for the diaphragm, whereas calf muscles show no pathological changes. CONCLUSION: SMA-related alterations appear to occur only when the muscles have reached a certain developmental maturity. Moreover, glial cells, in particular PSCs, play an important role in SMA pathogenesis.


Asunto(s)
Progresión de la Enfermedad , Desarrollo de Músculos , Músculo Esquelético/ultraestructura , Atrofia Muscular Espinal/patología , Unión Neuromuscular/ultraestructura , Animales , Diafragma/ultraestructura , Modelos Animales de Enfermedad , Femenino , Músculos Intercostales/ultraestructura , Ratones
15.
RNA Biol ; 11(11): 1430-46, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25692239

RESUMEN

Spinal Muscular Atrophy (SMA) is caused by deletions or mutations in the Survival Motor Neuron 1 (SMN1) gene. The second gene copy, SMN2, produces some, but not enough, functional SMN protein. SMN is essential to assemble small nuclear ribonucleoproteins (snRNPs) that form the spliceosome. However, it is not clear whether SMA is caused by defects in this function that could lead to splicing changes in all tissues, or by the impairment of an additional, less well characterized, but motoneuron-specific SMN function. We addressed the first possibility by exon junction microarray analysis of motoneurons (MNs) isolated by laser capture microdissection from a severe SMA mouse model. This revealed changes in multiple U2-dependent splicing events. Moreover, splicing appeared to be more strongly affected in MNs than in other cells. By testing mutiple genes in a model of progressive SMN depletion in NB2a neuroblastoma cells, we obtained evidence that U2-dependent splicing changes occur earlier than U12-dependent ones. As several of these changes affect genes coding for splicing regulators, this may acerbate the splicing response induced by low SMN levels and induce secondary waves of splicing alterations.


Asunto(s)
Regulación de la Expresión Génica , Neuronas Motoras/metabolismo , Empalme del ARN , Proteínas del Complejo SMN/genética , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Humanos , Intrones/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Interferencia de ARN , Factores de Empalme de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas del Complejo SMN/metabolismo , Septinas/genética , Septinas/metabolismo
16.
Methods Mol Biol ; 867: 325-47, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22454071

RESUMEN

Many inherited diseases are associated with changed splicing patterns, and alternative splicing influences several biological processes as well as the replication of certain viral pathogens. For this reason, there is a broad interest in modulating individual splicing events for therapeutic purposes. Based on the small nuclear RNA (snRNA) U7, we have developed expression vectors for short antisense RNAs that accumulate in the cell nucleus where splicing occurs and that can very specifically modulate the splicing of individual exons. More specifically, in the context of the fatal neuromuscular disorder Spinal Muscular Atrophy (SMA), we have shown that U7 snRNA constructs can restore the inclusion of exon 7 in the SMN2 gene and thereby alleviate or even fully cure disease symptoms in a severe mouse model for SMA. Here we describe more generally procedures to produce U7 constructs to induce exon inclusion and to test their efficiency in cell culture experiments at the level of RNA as well as protein. The analytical methods comprise reverse transcription (RT-)PCR to detect the splicing changes, quantitative real-time RT-PCR to measure U7 snRNA expression levels and western blot and immunofluorescence methods to detect a restoration of protein expression. Additionally, we indicate how U7 cassettes can be introduced into gene transfer vectors for in vivo experiments in animal models or to transduce cell systems that are not readily amenable to DNA transfection.


Asunto(s)
Elementos sin Sentido (Genética)/genética , Clonación Molecular/métodos , Exones , ARN Nuclear Pequeño/genética , Animales , Secuencia de Bases , Western Blotting/métodos , Terapia Genética/métodos , Lentivirus/genética , Ratones , Datos de Secuencia Molecular , Plásmidos/genética , Reacción en Cadena de la Polimerasa/métodos , ARN/genética , ARN/aislamiento & purificación , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Transfección
17.
Wiley Interdiscip Rev RNA ; 2(1): 79-91, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21956970

RESUMEN

The formation of defined 3(') ends is an important step in the biogenesis of mRNAs. In eukaryotic cells, all mRNA 3(') ends are generated by endonucleolytic cleavage of primary transcripts in reactions that are essentially posttranscriptional. Nevertheless, 3(') end formation is tightly connected to transcription in vivo, and a link with mRNA export to the cytoplasm has been postulated. Here, we briefly review the current knowledge about the two types of mRNA 3(') end processing reactions, cleavage/polyadenylation and histone RNA processing. We then focus on factors shared between these two reactions. In particular, we discuss evidence for new functions of the mammalian cleavage factor I subunit CF I(m) 68 in histone RNA 3(') processing and in the export of mature mRNAs from the nucleus to the cytoplasm.


Asunto(s)
Mamíferos/genética , Procesamiento de Término de ARN 3'/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Escisión y Poliadenilación de ARNm/fisiología , Animales , Histonas/genética , Humanos , Mamíferos/metabolismo , Modelos Biológicos , Factores de Escisión y Poliadenilación de ARNm/genética , Factores de Escisión y Poliadenilación de ARNm/metabolismo
18.
Mol Biol Cell ; 22(1): 91-104, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21119002

RESUMEN

Cleavage/polyadenylation of mRNAs and 3' processing of replication-dependent histone transcripts are both mediated by large complexes that share several protein components. Functional studies of these shared proteins are complicated by the cooperative binding of the individual subunits. For CstF-64, an additional difficulty is that symplekin and CstF-77 bind mutually exclusively to its hinge domain. Here we have identified CstF-64 and symplekin mutants that allowed us to distinguish between these interactions and to elucidate the role of CstF-64 in the two processing reactions. The interaction of CstF-64 with symplekin is limiting for histone RNA 3' processing but relatively unimportant for cleavage/polyadenylation. In contrast, the nuclear accumulation of CstF-64 depends on its binding to CstF-77 and not to symplekin. Moreover, the CstF-64 paralogue CstF-64Tau can compensate for the loss of CstF-64. As CstF-64Tau has a lower affinity for CstF-77 than CstF-64 and is relatively unstable, it is the minor form. However, it may become up-regulated when the CstF-64 level decreases, which has biological implications for spermatogenesis and probably also for other regulatory events. Thus, the interactions between CstF-64/CstF-64Tau and CstF-77 are important for the maintenance of stoichiometric nuclear levels of the CstF complex components and for their intracellular localization, stability, and function.


Asunto(s)
Factor de Estimulación del Desdoblamiento/química , Factor de Estimulación del Desdoblamiento/metabolismo , Proteínas Nucleares/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Far-Western Blotting , Factor de Estimulación del Desdoblamiento/genética , Técnica del Anticuerpo Fluorescente , Expresión Génica , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Poliadenilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Procesamiento de Término de ARN 3' , Señales de Poliadenilación de ARN 3' , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Escisión y Poliadenilación de ARNm/genética
20.
Neuromuscul Disord ; 20(11): 744-52, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20832308

RESUMEN

In Spinal Muscular Atrophy (SMA), the SMN1 gene is deleted or inactivated. Because of a splicing problem, the second copy gene, SMN2, generates insufficient amounts of functional SMN protein, leading to the death of spinal cord motoneurons. For a "severe" mouse SMA model (Smn -/-, hSMN2 +/+; with affected pups dying at 5-7 days), which most closely mimicks the genetic set-up in human SMA patients, we characterise SMA-related ultrastructural changes in neuromuscular junctions (NMJs) of two striated muscles with discrete functions. In the diaphragm, but not the soleus muscle of 4-days old SMA mice, mitochondria on both sides of the NMJs degenerate, and perisynaptic Schwann cells as well as endoneurial fibroblasts show striking changes in morphology. Importantly, NMJs of SMA mice in which a modified U7 snRNA corrects SMN2 splicing and delays or prevents SMA symptoms are normal. This ultrastructural study reveals novel features of NMJ alterations - in particular the involvement of perisynaptic Schwann cells - that may be relevant for human SMA pathogenesis.


Asunto(s)
Diafragma/ultraestructura , Atrofia Muscular Espinal/patología , Unión Neuromuscular/ultraestructura , Animales , Diafragma/patología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Mitocondrias/diagnóstico por imagen , Mitocondrias/patología , Neuronas Motoras/patología , Neuronas Motoras/ultraestructura , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/patología , Atrofia Muscular Espinal/genética , Unión Neuromuscular/genética , Unión Neuromuscular/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Ultrasonografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...