Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(3): 101682, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35124003

RESUMEN

miRNAs are short noncoding RNA molecules that regulate gene expression by inhibiting translation or inducing degradation of target mRNAs. miRNAs are often expressed as polycistronic transcripts, so-called miRNA clusters, containing several miRNA precursors. The largest mammalian miRNA cluster, the miR-379-410 cluster, is expressed primarily during embryonic development and in the adult brain; however, downstream regulation of this cluster is not well understood. Here, we investigated adenosine deamination to inosine (RNA editing) in the miR-379-410 cluster by adenosine deaminase acting on RNA (ADAR) enzymes as a possible mechanism modulating the expression and activity of these miRNAs in a brain-specific manner. We show that the levels of editing in the majority of mature miRNAs are lower than the editing levels of the corresponding site in primary miRNA precursors. However, for one miRNA, miR-376b-3p, editing was significantly higher in the mature form than in the primary precursor. We found miR-376b-3p maturation is negatively regulated by ADAR2 in an editing activity-independent manner, whereas ADAR1-mediated and ADAR2-mediated editing were observed to be competitive. In addition, the edited miR-376b-3p targets a different set of mRNAs than unedited miR-376b-3p, including 4-aminobutyrate aminotransferase, encoding the enzyme responsible for the catabolism of the neurotransmitter gamma aminobutyric acid (GABA). Expression of edited miR-376b-3p led to increased intracellular GABA levels as well as increased cell surface presentation of GABA type A receptors. Our results indicate that both editing and editing-independent effects modulate the expression of miR-376b-3p, with the potential to regulate GABAergic signaling in the brain.


Asunto(s)
MicroARNs , Proteínas de Unión al ARN , Ácido gamma-Aminobutírico , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Mamíferos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neurotransmisores , Edición de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ácido gamma-Aminobutírico/metabolismo
2.
Mol Cell ; 58(5): 870-85, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25921068

RESUMEN

Circular RNAs (circRNAs) are an endogenous class of animal RNAs. Despite their abundance, their function and expression in the nervous system are unknown. Therefore, we sequenced RNA from different brain regions, primary neurons, isolated synapses, as well as during neuronal differentiation. Using these and other available data, we discovered and analyzed thousands of neuronal human and mouse circRNAs. circRNAs were extraordinarily enriched in the mammalian brain, well conserved in sequence, often expressed as circRNAs in both human and mouse, and sometimes even detected in Drosophila brains. circRNAs were overall upregulated during neuronal differentiation, highly enriched in synapses, and often differentially expressed compared to their mRNA isoforms. circRNA expression correlated negatively with expression of the RNA-editing enzyme ADAR1. Knockdown of ADAR1 induced elevated circRNA expression. Together, we provide a circRNA brain expression atlas and evidence for important circRNA functions and values as biomarkers.


Asunto(s)
Encéfalo/metabolismo , ARN/metabolismo , Animales , Secuencia de Bases , Línea Celular , Drosophila melanogaster , Humanos , Ratones , Datos de Secuencia Molecular , Neurogénesis , Especificidad de Órganos , ARN/genética , ARN Circular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Sinapsis/metabolismo
3.
Nucleic Acids Res ; 47(4): e22, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30590609

RESUMEN

Adenosine to inosine editing is common in the human transcriptome and changes of this essential activity is associated with disease. Children with ADAR1 mutations develop fatal Aicardi-Goutières syndrome characterized by aberrant interferon expression. In contrast, ADAR1 overexpression is associated with increased malignancy of breast, lung and liver cancer. ADAR1 silencing in breast cancer cells leads to increased apoptosis, suggesting an anti-apoptotic function that promotes cancer progression. Yet, suitable high-throughput editing assays are needed to efficiently screen chemical libraries for modifiers of ADAR1 activity. We describe the development of a bioluminescent reporter system that facilitates rapid and accurate determination of endogenous editing activity. The system is based on the highly sensitive and quantitative Nanoluciferase that is conditionally expressed upon reporter-transcript editing. Stably introduced into cancer cell lines, the system reports on elevated endogenous ADAR1 editing activity induced by interferon as well as knockdown of ADAR1 and ADAR2. In a single-well setup we used the reporter in HeLa cells to screen a small molecule library of 33 000 compounds. This yielded a primary hit rate of 0.9% at 70% inhibition of editing. Thus, we provide a key tool for high-throughput identification of modifiers of A-to-I editing activity in cancer cells.


Asunto(s)
Adenosina Desaminasa/genética , Ensayos Analíticos de Alto Rendimiento , Neoplasias/genética , Proteínas de Unión al ARN/genética , Adenosina/genética , Apoptosis/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/patología , Edición Génica/métodos , Genes Reporteros/genética , Células HeLa , Humanos , Inosina/genética , Interferones/genética , Luciferasas/genética , Mediciones Luminiscentes/métodos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Bibliotecas de Moléculas Pequeñas/química , Transcriptoma/genética
4.
BMC Health Serv Res ; 21(1): 1207, 2021 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-34742302

RESUMEN

BACKGROUND: Failure to identify severely ill obstetric patients seeking acute care, and hence delaying treatment, can lead to maternal morbidity and mortality. Triage is the prioritization of patients seeking emergency care, based on clinical decision-making tools assessing medical urgency. While triage has been applied in general emergency medicine for 30 years, there are only a few obstetric triage systems (OTS) and obstetric triage has hitherto been unknown in Sweden. Obstetric triage is more complex than general triage since both mother and fetus require assessment, and pregnancy-related physiological changes must be taken into account. This paper aims to describe the development and an initial evaluation of the first OTS in Sweden. METHODS: A multidisciplinary team surveyed reasons to seek acute obstetric care and the current patient flow at the largest obstetric unit in Scandinavia, Sahlgrenska University Hospital, Gothenburg, Sweden, with about 10,000 deliveries/year. A semi-structured literature review on obstetric triage was undertaken. Based on the survey and the literature review the first Swedish OTS was developed and implemented. Patient satisfaction was followed by electronical questionnaires. Initial validity evaluation was performed, defined by the system's ability to identify patients with need for hospital admission, stratified by acuity level. RESULTS: The Gothenburg Obstetrical Triage System (GOTS) addresses the patient to one of five acuity levels based on both vital signs and 14 chief complaint algorithms. It entails recommendations for initial procedures of care as well as an acuity form for documentation. Initial evaluation of the system indicates good correlation between need for admission and acuity level. The implementation has provided the staff with an improved medical overview of the patients and patient flow and enabled the unit to monitor emergency care in a structured way. Implementation came along with increased patient and staff satisfaction. CONCLUSION: The GOTS is the first OTS developed in and for Sweden and implementation has improved management of obstetric patients seeking acute care. Patients are now prioritized according to level of acuity and the time to assessment and treatment of severely ill patients can be structurally evaluated. Both patients and staff express improved satisfaction with obstetric triage.


Asunto(s)
Servicios Médicos de Urgencia , Obstetricia , Servicio de Urgencia en Hospital , Femenino , Humanos , Embarazo , Suecia/epidemiología , Triaje
5.
BMC Biol ; 18(1): 6, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31937309

RESUMEN

BACKGROUND: Adenosine-to-inosine (A-to-I) RNA editing is a process that contributes to the diversification of proteins that has been shown to be essential for neurotransmission and other neuronal functions. However, the spatiotemporal and diversification properties of RNA editing in the brain are largely unknown. Here, we applied in situ sequencing to distinguish between edited and unedited transcripts in distinct regions of the mouse brain at four developmental stages, and investigate the diversity of the RNA landscape. RESULTS: We analyzed RNA editing at codon-altering sites using in situ sequencing at single-cell resolution, in combination with the detection of individual ADAR enzymes and specific cell type marker transcripts. This approach revealed cell-type-specific regulation of RNA editing of a set of transcripts, and developmental and regional variation in editing levels for many of the targeted sites. We found increasing editing diversity throughout development, which arises through regional- and cell type-specific regulation of ADAR enzymes and target transcripts. CONCLUSIONS: Our single-cell in situ sequencing method has proved useful to study the complex landscape of RNA editing and our results indicate that this complexity arises due to distinct mechanisms of regulating individual RNA editing sites, acting both regionally and in specific cell types.


Asunto(s)
Encéfalo/metabolismo , Edición de ARN , Adenosina/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Inosina/metabolismo , Ratones , Análisis de Secuencia de ARN , Análisis Espacio-Temporal
6.
Semin Cell Dev Biol ; 79: 123-130, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29146145

RESUMEN

Cancer arises when pathways that control cell functions such as proliferation and migration are dysregulated to such an extent that cells start to divide uncontrollably and eventually spread throughout the body, ultimately endangering the survival of an affected individual. It is well established that somatic mutations are important in cancer initiation and progression as well as in creation of tumor diversity. Now also modifications of the transcriptome are emerging as a significant force during the transition from normal cell to malignant tumor. Editing of adenosine (A) to inosine (I) in double-stranded RNA, catalyzed by adenosine deaminases acting on RNA (ADARs), is one dynamic modification that in a combinatorial manner can give rise to a very diverse transcriptome. Since the cell interprets inosine as guanosine (G), editing can result in non-synonymous codon changes in transcripts as well as yield alternative splicing, but also affect targeting and disrupt maturation of microRNA. ADAR editing is essential for survival in mammals but its dysregulation can lead to cancer. ADAR1 is for instance overexpressed in, e.g., lung cancer, liver cancer, esophageal cancer and chronic myoelogenous leukemia, which with few exceptions promotes cancer progression. In contrast, ADAR2 is lowly expressed in e.g. glioblastoma, where the lower levels of ADAR2 editing leads to malignant phenotypes. Altogether, RNA editing by the ADAR enzymes is a powerful regulatory mechanism during tumorigenesis. Depending on the cell type, cancer progression seems to mainly be induced by ADAR1 upregulation or ADAR2 downregulation, although in a few cases ADAR1 is instead downregulated. In this review, we discuss how aberrant editing of specific substrates contributes to malignancy.


Asunto(s)
Adenosina Desaminasa/metabolismo , Neoplasias/genética , Edición de ARN , ARN Bicatenario/genética , Proteínas de Unión al ARN/metabolismo , Animales , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Isoformas de ARN/genética , Isoformas de ARN/metabolismo , ARN Bicatenario/metabolismo
7.
Trends Genet ; 32(3): 165-175, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26803450

RESUMEN

Post-transcriptional RNA modification by adenosine to inosine (A-to-I) editing expands the functional output of many important neuronally expressed genes. The mechanism provides flexibility in the proteome by expanding the variety of isoforms, and is a requisite for neuronal function. Indeed, targets for editing include key mediators of synaptic transmission with an overall significant effect on neuronal signaling. In addition, editing influences splice-site choice and miRNA targeting capacity, and thereby regulates neuronal gene expression. Editing efficiency at most of these sites increases during neuronal differentiation and brain maturation in a spatiotemporal manner. This editing-induced dynamics in the transcriptome is essential for normal brain development, and we are only beginning to understand its role in neuronal function. In this review we discuss the impact of RNA editing in the brain, with special emphasis on the physiological consequences for neuronal development and plasticity.


Asunto(s)
Encéfalo/citología , Neuronas/citología , Edición de ARN , Animales , Mamíferos , Empalme del ARN , Transcripción Genética
8.
J Cell Sci ; 130(4): 745-753, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28082424

RESUMEN

Adenosine to inosine (A-to-I) RNA editing is important for a functional brain, and most known sites that are subject to selective RNA editing have been found to result in diversified protein isoforms that are involved in neurotransmission. In the absence of the active editing enzymes ADAR1 or ADAR2 (also known as ADAR and ADARB1, respectively), mice fail to survive until adulthood. Nuclear A-to-I editing of neuronal transcripts is regulated during brain development, with low levels of editing in the embryo and a dramatic increase after birth. Yet, little is known about the mechanisms that regulate editing during development. Here, we demonstrate lower levels of ADAR2 in the nucleus of immature neurons than in mature neurons. We show that importin-α4 (encoded by Kpna3), which increases during neuronal maturation, interacts with ADAR2 and contributes to the editing efficiency by bringing it into the nucleus. Moreover, we detect an increased number of interactions between ADAR2 and the nuclear isomerase Pin1 as neurons mature, which contribute to ADAR2 protein stability. Together, these findings explain how the nuclear editing of substrates that are important for neuronal function can increase as the brain develops.


Asunto(s)
Adenosina Desaminasa/metabolismo , Adenosina/metabolismo , Núcleo Celular/metabolismo , Inosina/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Edición de ARN , Adenosina Desaminasa/química , Animales , Diferenciación Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Células HEK293 , Humanos , Ratones , Modelos Biológicos , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Señales de Localización Nuclear/metabolismo , Unión Proteica , Ratas , alfa Carioferinas
9.
Nucleic Acids Res ; 45(7): 4189-4201, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28053121

RESUMEN

Adenosine deaminases acting on RNA (ADARs) catalyze the editing of adenosine residues to inosine (A-to-I) within RNA sequences, mostly in the introns and UTRs (un-translated regions). The significance of editing within non-coding regions of RNA is poorly understood. Here, we demonstrate that association of ADAR2 with RNA stabilizes a subset of transcripts. ADAR2 interacts with and edits the 3΄UTR of nuclear-retained Cat2 transcribed nuclear RNA (Ctn RNA). In absence of ADAR2, the abundance and half-life of Ctn RNA are significantly reduced. Furthermore, ADAR2-mediated stabilization of Ctn RNA occurred in an editing-independent manner. Unedited Ctn RNA shows enhanced interaction with the RNA-binding proteins HuR and PARN [Poly(A) specific ribonuclease deadenylase]. HuR and PARN destabilize Ctn RNA in absence of ADAR2, indicating that ADAR2 stabilizes Ctn RNA by antagonizing its degradation by PARN and HuR. Transcriptomic analysis identified other RNAs that are regulated by a similar mechanism. In summary, we identify a regulatory mechanism whereby ADAR2 enhances target RNA stability by limiting the interaction of RNA-destabilizing proteins with their cognate substrates.


Asunto(s)
Adenosina Desaminasa/metabolismo , Proteína 1 Similar a ELAV/metabolismo , Exorribonucleasas/metabolismo , Estabilidad del ARN , Proteínas de Unión al ARN/metabolismo , Regiones no Traducidas 3' , Animales , Transportador de Aminoácidos Catiônicos 2/genética , Transportador de Aminoácidos Catiônicos 2/metabolismo , Ratones , Edición de ARN , ARN Largo no Codificante/metabolismo
10.
RNA Biol ; 15(6): 829-831, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29671387

RESUMEN

The genetic alphabet consists of the four letters: C, A, G, and T in DNA and C,A,G, and U in RNA. Triplets of these four letters jointly encode 20 different amino acids out of which proteins of all organisms are built. This system is universal and is found in all kingdoms of life. However, bases in DNA and RNA can be chemically modified. In DNA, around 10 different modifications are known, and those have been studied intensively over the past 20 years. Scientific studies on DNA modifications and proteins that recognize them gave rise to the large field of epigenetic and epigenomic research. The outcome of this intense research field is the discovery that development, ageing, and stem-cell dependent regeneration but also several diseases including cancer are largely controlled by the epigenetic state of cells. Consequently, this research has already led to the first FDA approved drugs that exploit the gained knowledge to combat disease. In recent years, the ~150 modifications found in RNA have come to the focus of intense research. Here we provide a perspective on necessary and expected developments in the fast expanding area of RNA modifications, termed epitranscriptomics.


Asunto(s)
ADN de Neoplasias , Epigénesis Genética , Epigenómica/normas , Perfilación de la Expresión Génica/normas , Regulación Neoplásica de la Expresión Génica , Neoplasias , ARN Neoplásico , Transcriptoma , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Europa (Continente) , Perfilación de la Expresión Génica/métodos , Humanos , Neoplasias/genética , Neoplasias/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo
11.
Cell Mol Life Sci ; 72(21): 4063-76, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26223268

RESUMEN

The human genome is under constant invasion by retrotransposable elements. The most successful of these are the Alu elements; with a copy number of over a million, they occupy about 10 % of the entire genome. Interestingly, the vast majority of these Alu insertions are located in gene-rich regions, and one-third of all human genes contains an Alu insertion. Alu sequences are often embedded in gene sequence encoding pre-mRNAs and mature mRNAs, usually as part of their intron or UTRs. Once transcribed, they can regulate gene expression as well as increase the number of RNA isoforms expressed in a tissue or a species. They also regulate the function of other RNAs, like microRNAs, circular RNAs, and potentially long non-coding RNAs. Mechanistically, Alu elements exert their effects by influencing diverse processes, such as RNA editing, exonization, and RNA processing. In so doing, they have undoubtedly had a profound effect on human evolution.


Asunto(s)
Elementos Alu/fisiología , Edición de ARN , Procesamiento Postranscripcional del ARN , Animales , Apoptosis/genética , Evolución Molecular , Exones , Regulación de la Expresión Génica , Genoma Humano , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Primates/genética , ARN/metabolismo , ARN Circular , Regiones no Traducidas
12.
Genome Res ; 22(8): 1477-87, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22645261

RESUMEN

Adenosine-to-inosine (A-to-I) RNA editing targets double-stranded RNA stem-loop structures in the mammalian brain. It has previously been shown that miRNAs are substrates for A-to-I editing. For the first time, we show that for several definitions of edited miRNA, the level of editing increases with development, thereby indicating a regulatory role for editing during brain maturation. We use high-throughput RNA sequencing to determine editing levels in mature miRNA, from the mouse transcriptome, and compare these with the levels of editing in pri-miRNA. We show that increased editing during development gradually changes the proportions of the two miR-376a isoforms, which previously have been shown to have different targets. Several other miRNAs that also are edited in the seed sequence show an increased level of editing through development. By comparing editing of pri-miRNA with editing and expression of the corresponding mature miRNA, we also show an editing-induced developmental regulation of miRNA expression. Taken together, our results imply that RNA editing influences the miRNA repertoire during brain maturation.


Asunto(s)
Adenosina/metabolismo , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Inosina/metabolismo , MicroARNs/metabolismo , Edición de ARN , Adenosina/genética , Animales , Secuencia de Bases , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Biología Computacional , Dendritas/genética , Dendritas/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Inosina/genética , Ratones , MicroARNs/genética , Isoformas de ARN/genética , Isoformas de ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcriptoma
13.
Hum Mol Genet ; 21(2): 311-21, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21984433

RESUMEN

Schizophrenia and bipolar disorder (BPD) are common neurodevelopmental disorders, characterized by various life-crippling symptoms and high suicide rates. Multiple studies support a strong genetic involvement in the etiology of these disorders, although patterns of inheritance are variable and complex. Adenosine-to-inosine RNA editing is a cellular mechanism, which has been implicated in mental disorders and suicide. To examine the involvement of altered RNA editing in these disorders, we: (i) quantified the mRNA levels of the adenosine deaminase acting on RNA (ADAR) editing enzymes by real-time quantitative polymerase chain reaction, and (ii) measured the editing levels in transcripts of several neuroreceptors using 454 high-throughput sequencing, in dorsolateral-prefrontal cortices of schizophrenics, BPD patients and controls. Increased expression of specific ADAR2 variants with diminished catalytic activity was observed in schizophrenia. Our results also indicate that the I/V editing site in the glutamate receptor, ionotropic kainate 2 (GRIK2) transcript is under-edited in BPD (type I) patients (45.8 versus 53.9%, P= 0.023). GRIK2 has been implicated in mood disorders, and editing of its I/V site can modulate Ca(+2) permeability of the channel, consistent with numerous observations of elevated intracellular Ca(+2) levels in BPD patients. Our findings may therefore, at least partly, explain a molecular mechanism underlying the disorder. In addition, an intriguing correlation was found between editing events on separate exons of GRIK2. Finally, multiple novel editing sites were detected near previously known sites, albeit most with very low editing rates. This supports the hypothesis raised previously regarding the existence of wide-spread low-level 'background' editing as a mechanism that enhances adaptation and evolvability.


Asunto(s)
Trastornos Mentales/genética , Edición de ARN , Humanos , Receptores de Ácido Kaínico/genética , Receptor de Ácido Kaínico GluK2
14.
Nucleic Acids Res ; 40(19): 9876-86, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22848101

RESUMEN

Transcripts have been found to be site selectively edited from adenosine-to-inosine (A-to-I) in the mammalian brain, mostly in genes involved in neurotransmission. While A-to-I editing occurs at double-stranded structures, other structural requirements are largely unknown. We have investigated the requirements for editing at the I/M site in the Gabra-3 transcript of the GABA(A) receptor. We identify an evolutionarily conserved intronic duplex, 150 nt downstream of the exonic hairpin where the I/M site resides, which is required for its editing. This is the first time a distant RNA structure has been shown to be important for A-to-I editing. We demonstrate that the element also can induce editing in related but normally not edited RNA sequences. In human, thousands of genes are edited in duplexes formed by inverted repeats in non-coding regions. It is likely that numerous such duplexes can induce editing of coding regions throughout the transcriptome.


Asunto(s)
Intrones , Edición de ARN , Secuencias Reguladoras de Ácido Ribonucleico , Animales , Encéfalo/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Conformación de Ácido Nucleico , Empalme del ARN , Ratas , Receptores de GABA-A/genética , Porcinos
15.
Arthritis Rheum ; 64(5): 1579-88, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22131101

RESUMEN

OBJECTIVE: To assess the efficacy of paquinimod, a new immunomodulatory small molecule, in a murine lupus model, and to evaluate its pharmacokinetics and tolerability in systemic lupus erythematosus (SLE) patients at doses predicted to be efficacious and safe and determine the maximum tolerated dose. METHODS: The efficacy of paquinimod was studied in lupus-prone MRL-lpr/lpr mice and compared with that of established SLE treatments. Dose-response data and pharmacokinetic data were used to calculate effective and safe clinical doses of paquinimod. The pharmacokinetics and tolerability of paquinimod were evaluated in a phase Ib double-blind, placebo controlled, dose-ranging study in which cohorts of SLE patients received daily oral treatment for 12 weeks. RESULTS: Paquinimod treatment resulted in disease inhibition in MRL-lpr/lpr mice, comparable to that obtained with prednisolone and mycophenolate mofetil; prominent effects on disease manifestations and serologic markers and a steroid-sparing effect were observed. In patients with SLE, the pharmacokinetic properties of paquinimod were linear and well suitable for once-daily oral treatment. The majority of the adverse events (AEs) were mild or moderate, and transient. The most frequent AEs were arthralgia and myalgia, reported with the highest dose levels of paquinimod (4.5 mg/day and 6.0 mg/day). At the 4.5 mg/day dose level and higher, some AEs of severe intensity and serious adverse events were reported. CONCLUSION: Paquinimod effectively inhibited disease and had a steroid-sparing effect in experimental lupus. Results from preclinical models together with pharmacokinetic data were successfully translated into a safe clinical dose range, and doses of up to 3.0 mg/day were well tolerated in the SLE patients. Taken together, the promising combined data from a murine model and human SLE support the future clinical development of paquinimod.


Asunto(s)
Inmunosupresores , Lupus Eritematoso Sistémico/tratamiento farmacológico , Quinolinas/química , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Método Doble Ciego , Femenino , Humanos , Inmunosupresores/efectos adversos , Inmunosupresores/farmacocinética , Inmunosupresores/uso terapéutico , Riñón/metabolismo , Riñón/patología , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Masculino , Ratones , Ratones Endogámicos MRL lpr , Persona de Mediana Edad , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapéutico , Prednisolona/uso terapéutico , Quinolinas/efectos adversos , Quinolinas/farmacocinética , Quinolinas/uso terapéutico , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven
16.
J Biol Chem ; 286(3): 2031-40, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21030585

RESUMEN

Recoding by adenosine-to-inosine RNA editing plays an important role in diversifying proteins involved in neurotransmission. We have previously shown that the Gabra-3 transcript, coding for the α3 subunit of the GABA(A) receptor is edited in mouse, causing an isoleucine to methionine (I/M) change. Here we show that this editing event is evolutionarily conserved from human to chicken. Analyzing recombinant GABA(A) receptor subunits expressed in HEK293 cells, our results suggest that editing at the I/M site in α3 has functional consequences on receptor expression. We demonstrate that I/M editing reduces the cell surface and the total number of α3 subunits. The reduction in cell surface levels is independent of the subunit combination as it is observed for α3 in combination with either the ß2 or the ß3 subunit. Further, an amino acid substitution at the corresponding I/M site in the α1 subunit has a similar effect on cell surface presentation, indicating the importance of this site for receptor trafficking. We show that the I/M editing during brain development is inversely related to the α3 protein abundance. Our results suggest that editing controls trafficking of α3-containing receptors and may therefore facilitate the switch of subunit compositions during development as well as the subcellular distribution of α subunits in the adult brain.


Asunto(s)
Expresión Génica , Edición de ARN/fisiología , Receptores de GABA-A/biosíntesis , Proteínas Recombinantes/biosíntesis , Adenosina/genética , Adenosina/metabolismo , Adulto , Sustitución de Aminoácidos , Animales , Encéfalo/metabolismo , Células HEK293 , Humanos , Inosina/genética , Inosina/metabolismo , Ratones , Transporte de Proteínas/fisiología , Ratas , Receptores de GABA-A/genética , Proteínas Recombinantes/genética
17.
18.
RNA Biol ; 9(8): 1054-65, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22858680

RESUMEN

Editing by ADAR enzymes is essential for mammalian life. Still, knowledge of the spatio-temporal editing patterns in mammals is limited. By use of 454 amplicon sequencing we examined the editing status of 12 regionally extracted mRNAs from porcine developing brain encompassing a total of 64 putative ADAR editing sites. In total 24 brain tissues, dissected from up to five regions from embryonic gestation day 23, 42, 60, 80, 100 and 115, were examined for editing.   Generally, editing increased during embryonic development concomitantly with an increase in ADAR2 mRNA level. Notably, the Gria2 (GluR-B) Q/R site, reported to be ~100% edited in previous studies, is only 54% edited at embryonic day 23. Transcripts with multiple editing sites in close proximity to each other exhibit coupled editing and an extraordinary incidence of long-range coupling of editing events more than 32 kb apart is observed for the kainate glutamate receptor 2 transcript, Grik2. Our study reveals complex spatio-temporal ADAR editing patterns of coordinated editing events that may play important roles in the development of the mammalian brain.


Asunto(s)
Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Edición de ARN , Sus scrofa/embriología , Animales , Humanos , Ratones , Análisis de Secuencia de ADN , Sus scrofa/metabolismo
19.
Nucleic Acids Res ; 37(20): 6916-26, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19740768

RESUMEN

Adenosine-to-inosine (A-to-I) editing has been shown to be an important mechanism that increases protein diversity in the brain of organisms from human to fly. The family of ADAR enzymes converts some adenosines of RNA duplexes to inosines through hydrolytic deamination. The adenosine recognition mechanism is still largely unknown. Here, to investigate it, we analyzed a set of selectively edited substrates with a cluster of edited sites. We used a large set of individual transcripts sequenced by the 454 sequencing technique. On average, we analyzed 570 single transcripts per edited region at four different developmental stages from embryogenesis to adulthood. To our knowledge, this is the first time, large-scale sequencing has been used to determine synchronous editing events. We demonstrate that edited sites are only coupled within specific distances from each other. Furthermore, our results show that the coupled sites of editing are positioned on the same side of a helix, indicating that the three-dimensional structure is key in ADAR enzyme substrate recognition. Finally, we propose that editing by the ADAR enzymes is initiated by their attraction to one principal site in the substrate.


Asunto(s)
Adenosina/metabolismo , Inosina/metabolismo , Edición de ARN , ARN/química , Animales , Secuencia de Bases , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN/metabolismo
20.
BMC Bioinformatics ; 11: 6, 2010 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-20047656

RESUMEN

BACKGROUND: Several bioinformatic approaches have previously been used to find novel sites of ADAR mediated A-to-I RNA editing in human. These studies have discovered thousands of genes that are hyper-edited in their non-coding intronic regions, especially in alu retrotransposable elements, but very few substrates that are site-selectively edited in coding regions. Known RNA edited substrates suggest, however, that site selective A-to-I editing is particularly important for normal brain development in mammals. RESULTS: We have compiled a screen that enables the identification of new sites of site-selective editing, primarily in coding sequences. To avoid hyper-edited repeat regions, we applied our screen to the alu-free mouse genome. Focusing on the mouse also facilitated better experimental verification. To identify candidate sites of RNA editing, we first performed an explorative screen based on RNA structure and genomic sequence conservation. We further evaluated the results of the explorative screen by determining which transcripts were enriched for A-G mismatches between the genomic template and the expressed sequence since the editing product, inosine (I), is read as guanosine (G) by the translational machinery. For expressed sequences, we only considered coding regions to focus entirely on re-coding events. Lastly, we refined the results from the explorative screen using a novel scoring scheme based on characteristics for known A-to-I edited sites. The extent of editing in the final candidate genes was verified using total RNA from mouse brain and 454 sequencing. CONCLUSIONS: Using this method, we identified and confirmed efficient editing at one site in the Gabra3 gene. Editing was also verified at several other novel sites within candidates predicted to be edited. Five of these sites are situated in genes coding for the neuron-specific RNA binding proteins HuB and HuD.


Asunto(s)
Adenosina/genética , Genoma , Inosina/genética , Neuronas/metabolismo , Edición de ARN , Proteínas de Unión al ARN/química , Adenosina/metabolismo , Elementos Alu/genética , Animales , Secuencia de Bases , Biología Computacional/métodos , Ratones , Datos de Secuencia Molecular , Filogenia , ARN/química , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA