Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Cell ; 56(2): 311-322, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25263597

RESUMEN

Inhibition of muscleblind-like (MBNL) activity due to sequestration by microsatellite expansion RNAs is a major pathogenic event in the RNA-mediated disease myotonic dystrophy (DM). Although MBNL1 and MBNL2 bind to nascent transcripts to regulate alternative splicing during muscle and brain development, another major binding site for the MBNL protein family is the 3' untranslated region of target RNAs. Here, we report that depletion of Mbnl proteins in mouse embryo fibroblasts leads to misregulation of thousands of alternative polyadenylation events. HITS-CLIP and minigene reporter analyses indicate that these polyadenylation switches are a direct consequence of MBNL binding to target RNAs. Misregulated alternative polyadenylation also occurs in skeletal muscle in a mouse polyCUG model and human DM, resulting in the persistence of neonatal polyadenylation patterns. These findings reveal an additional developmental function for MBNL proteins and demonstrate that DM is characterized by misregulation of pre-mRNA processing at multiple levels.


Asunto(s)
Empalme Alternativo/genética , Proteínas Portadoras/genética , Proteínas de Unión al ADN/genética , Poliadenilación/genética , Proteínas de Unión al ARN/genética , Regiones no Traducidas 3'/genética , Animales , Sitios de Unión/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Repeticiones de Microsatélite/genética , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Distrofia Miotónica/genética , Unión Proteica , Interferencia de ARN , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Proteínas de Unión al ARN/metabolismo
2.
Adv Exp Med Biol ; 825: 353-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25201111

RESUMEN

RNA-binding proteins (RBPs) play pivotal roles in multiple cellular pathways from transcription to RNA turnover by interacting with RNA sequence and/or structural elements to form distinct RNA-protein complexes. Since these complexes are required for the normal regulation of gene expression, mutations that alter RBP functions may result in a cascade of deleterious events that lead to severe disease. Here, we focus on a group of hereditary disorders, the microsatellite expansion diseases, which alter RBP activities and result in abnormal neurological and neuromuscular phenotypes. While many of these diseases are classified as adult-onset disorders, mounting evidence indicates that disruption of normal RNA-protein interaction networks during embryogenesis modifies developmental pathways, which ultimately leads to disease manifestations later in life. Efforts to understand the molecular basis of these disorders has already uncovered novel pathogenic mechanisms, including RNA toxicity and repeat-associated non-ATG (RAN) translation, and current studies suggest that additional surprising insights into cellular regulatory pathways will emerge in the future.


Asunto(s)
Expansión de las Repeticiones de ADN , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Repeticiones de Microsatélite , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Humanos
3.
Clin Transl Immunology ; 9(11): e1214, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33304583

RESUMEN

OBJECTIVES: Genetic or acquired defects in FOXP3+ regulatory T cells (Tregs) play a key role in many immune-mediated diseases including immune dysregulation polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome. Previously, we demonstrated CD4+ T cells from healthy donors and IPEX patients can be converted into functional Treg-like cells by lentiviral transfer of FOXP3 (CD4LVFOXP3). These CD4LVFOXP3 cells have potent regulatory function, suggesting their potential as an innovative therapeutic. Here, we present molecular and preclinical in vivo data supporting CD4LVFOXP3 cell clinical progression. METHODS: The molecular characterisation of CD4LVFOXP3 cells included flow cytometry, qPCR, RNA-seq and TCR-seq. The in vivo suppressive function of CD4LVFOXP3 cells was assessed in xenograft-versus-host disease (xeno-GvHD) and FOXP3-deficient IPEX-like humanised mouse models. The safety of CD4LVFOXP3 cells was evaluated using peripheral blood (PB) humanised (hu)- mice testing their impact on immune response against pathogens, and immune surveillance against tumor antigens. RESULTS: We demonstrate that the conversion of CD4+ T cells to CD4LVFOXP3 cells leads to specific transcriptional changes as compared to CD4+ T-cell transduction in the absence of FOXP3, including upregulation of Treg-related genes. Furthermore, we observe specific preservation of a polyclonal TCR repertoire during in vitro cell production. Both allogeneic and autologous CD4LVFOXP3 cells protect from xeno-GvHD after two sequential infusions of effector T cells. CD4LVFOXP3 cells prevent hyper-proliferation of CD4+ memory T cells in the FOXP3-deficient IPEX-like hu-mice. CD4LVFOXP3 cells do not impede in vivo expansion of antigen-primed T cells or tumor clearance in the PB hu-mice. CONCLUSION: These data support the clinical readiness of CD4LVFOXP3 cells to treat IPEX syndrome and other immune-mediated diseases caused by insufficient or dysfunctional FOXP3+ Tregs.

4.
Nat Commun ; 9(1): 2189, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29875359

RESUMEN

Alternative splicing (AS) is one crucial step of gene expression that must be tightly regulated during neurodevelopment. However, the precise timing of developmental splicing switches and the underlying regulatory mechanisms are poorly understood. Here we systematically analyze the temporal regulation of AS in a large number of transcriptome profiles of developing mouse cortices, in vivo purified neuronal subtypes, and neurons differentiated in vitro. Our analysis reveals early-switch and late-switch exons in genes with distinct functions, and these switches accurately define neuronal maturation stages. Integrative modeling suggests that these switches are under direct and combinatorial regulation by distinct sets of neuronal RNA-binding proteins including Nova, Rbfox, Mbnl, and Ptbp. Surprisingly, various neuronal subtypes in the sensory systems lack Nova and/or Rbfox expression. These neurons retain the "immature" splicing program in early-switch exons, affecting numerous synaptic genes. These results provide new insights into the organization and regulation of the neurodevelopmental transcriptome.


Asunto(s)
Empalme Alternativo , Sistema Nervioso Central/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/genética , Animales , Diferenciación Celular/genética , Sistema Nervioso Central/embriología , Sistema Nervioso Central/crecimiento & desarrollo , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Modelos Neurológicos , Neuronas/citología , Neuronas/metabolismo , Proteínas de Unión al ARN/genética , Factores de Tiempo
5.
Cell Rep ; 12(7): 1159-68, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26257173

RESUMEN

For some neurological disorders, disease is primarily RNA mediated due to expression of non-coding microsatellite expansion RNAs (RNA(exp)). Toxicity is thought to result from enhanced binding of proteins to these expansions and depletion from their normal cellular targets. However, experimental evidence for this sequestration model is lacking. Here, we use HITS-CLIP and pre-mRNA processing analysis of human control versus myotonic dystrophy (DM) brains to provide compelling evidence for this RNA toxicity model. MBNL2 binds directly to DM repeat expansions in the brain, resulting in depletion from its normal RNA targets with downstream effects on alternative splicing and polyadenylation. Similar RNA processing defects were detected in Mbnl compound-knockout mice, highlighted by dysregulation of Mapt splicing and fetal tau isoform expression in adults. These results demonstrate that MBNL proteins are directly sequestered by RNA(exp) in the DM brain and introduce a powerful experimental tool to evaluate RNA-mediated toxicity in other expansion diseases.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Distrofia Miotónica/genética , Empalme del ARN , ARN no Traducido/genética , Proteínas de Unión al ARN/metabolismo , Animales , Expansión de las Repeticiones de ADN , Proteínas de Unión al ADN/genética , Humanos , Ratones , Repeticiones de Microsatélite , Distrofia Miotónica/metabolismo , Proteínas de Unión al ARN/genética , Proteínas tau/genética , Proteínas tau/metabolismo
6.
Brain Res ; 1584: 3-14, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-24709120

RESUMEN

A novel RNA-mediated disease mechanism has emerged from studies on dominantly inherited neurological disorders caused by unstable microsatellite expansions in non-coding regions of the genome. These non-coding tandem repeat expansions trigger the production of unusual RNAs that gain a toxic function, which involves the formation of RNA repeat structures that interact with, and alter the activities of, various factors required for normal RNA processing as well as additional cellular functions. In this review, we explore the deleterious effects of toxic RNA expression and discuss the various model systems currently available for studying RNA gain-of-function in neurologic diseases. Common themes, including bidirectional transcription and repeat-associated non-ATG (RAN) translation, have recently emerged from expansion disease studies. These and other discoveries have highlighted the need for further investigations designed to provide the additional mechanistic insights essential for future therapeutic development.


Asunto(s)
Expansión de las Repeticiones de ADN , Repeticiones de Microsatélite/genética , Enfermedades del Sistema Nervioso/genética , Proteínas de Unión al ARN/genética , ARN/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Ataxia/genética , Proteína C9orf72 , Modelos Animales de Enfermedad , Síndrome del Cromosoma X Frágil/genética , Demencia Frontotemporal/genética , Humanos , Distrofia Miotónica/genética , Proteínas/genética , Temblor/genética
7.
Cell Reprogram ; 15(2): 166-77, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23550732

RESUMEN

Dystrophia myotonica type 1 (DM1) is an autosomal dominant multisystem disorder. The pathogenesis of central nervous system (CNS) involvement is poorly understood. Disease-specific induced pluripotent stem cell (iPSC) lines would provide an alternative model. In this study, we generated two DM1 lines and a normal iPSC line from dermal fibroblasts by retroviral transduction of Yamanaka's four factors (hOct4, hSox2, hKlf4, and hc-Myc). Both DM1 and control iPSC clones showed typical human embryonic stem cell (hESC) growth patterns with a high nuclear-to-cytoplasm ratio. The iPSC colonies maintained the same growth pattern through subsequent passages. All iPSC lines expressed stem cell markers and differentiated into cells derived from three embryonic germ layers. All iPSC lines underwent normal neural differentiation. Intranuclear RNA foci, a hallmark of DM1, were detected in DM1 iPSCs, neural stem cells (NSCs), and terminally differentiated neurons and astrocytes. In conclusion, we have successfully established disease-specific human DM1 iPSC lines, NSCs, and neuronal lineages with pathognomonic intranuclear RNA foci, which offer an unlimited cell resource for CNS mechanistic studies and a translational platform for therapeutic development.


Asunto(s)
Diferenciación Celular , Dermis/metabolismo , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Distrofia Miotónica/metabolismo , Células-Madre Neurales/metabolismo , Línea Celular , Dermis/patología , Femenino , Fibroblastos/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Persona de Mediana Edad , Distrofia Miotónica/patología , Distrofia Miotónica/terapia , Células-Madre Neurales/patología , Trasplante de Células Madre
8.
Peptides ; 34(1): 201-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21839791

RESUMEN

Allatotropin is an insect neuropeptide with pleiotropic actions on a variety of different tissues. In the present work we describe the identification, cloning and functional and molecular characterization of an Aedes aegypti allatotropin receptor (AeATr) and provide a detailed quantitative study of the expression of the AeATr gene in the adult mosquito. Analysis of the tissue distribution of AeATr mRNA in adult female revealed high transcript levels in the nervous system (brain, abdominal, thoracic and ventral ganglia), corpora allata-corpora cardiaca complex and ovary. The receptor is also expressed in heart, hindgut and male testis and accessory glands. Separation of the corpora allata (CA) and corpora cardiaca followed by analysis of gene expression in the isolated glands revealed expression of the AeATr primarily in the CA. In the female CA, the AeATr mRNA levels were low in the early pupae, started increasing 6h before adult eclosion and reached a maximum 24h after female emergence. Blood feeding resulted in a decrease in transcript levels. The pattern of changes of AeATr mRNA resembles the changes in JH biosynthesis. Fluorometric Imaging Plate Reader recordings of calcium transients in HEK293 cells expressing the AeATr showed a selective response to A. aegypti allatotropin stimulation in the low nanomolar concentration range. Our studies suggest that the AeATr play a role in the regulation of JH synthesis in mosquitoes.


Asunto(s)
Culicidae/metabolismo , Hormonas de Insectos/metabolismo , Proteínas de Insectos/metabolismo , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Corpora Allata/metabolismo , Femenino , Humanos , Inmunohistoquímica , Proteínas de Insectos/química , Proteínas de Insectos/clasificación , Proteínas de Insectos/genética , Hormonas Juveniles/química , Hormonas Juveniles/clasificación , Hormonas Juveniles/genética , Hormonas Juveniles/metabolismo , Masculino , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/clasificación , Receptores Acoplados a Proteínas G/genética
9.
Neuron ; 75(3): 437-50, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22884328

RESUMEN

The RNA-mediated disease model for myotonic dystrophy (DM) proposes that microsatellite C(C)TG expansions express toxic RNAs that disrupt splicing regulation by altering MBNL1 and CELF1 activities. While this model explains DM manifestations in muscle, less is known about the effects of C(C)UG expression on the brain. Here, we report that Mbnl2 knockout mice develop several DM-associated central nervous system (CNS) features including abnormal REM sleep propensity and deficits in spatial memory. Mbnl2 is prominently expressed in the hippocampus and Mbnl2 knockouts show a decrease in NMDA receptor (NMDAR) synaptic transmission and impaired hippocampal synaptic plasticity. While Mbnl2 loss did not significantly alter target transcript levels in the hippocampus, misregulated splicing of hundreds of exons was detected using splicing microarrays, RNA-seq, and HITS-CLIP. Importantly, the majority of the Mbnl2-regulated exons examined were similarly misregulated in DM. We propose that major pathological features of the DM brain result from disruption of the MBNL2-mediated developmental splicing program.


Asunto(s)
Empalme Alternativo , Encéfalo/fisiopatología , Distrofia Miotónica/genética , Proteínas de Unión al ARN/genética , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Distrofia Miotónica/metabolismo , Distrofia Miotónica/fisiopatología , Plasticidad Neuronal/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transmisión Sináptica/genética
10.
Peptides ; 31(3): 442-50, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19409436

RESUMEN

In the present work we describe the functional and molecular characterization of two Aedes aegypti allatostatin-C receptor paralogs (AeAS-CrA and AeAS-CrB) and provide a detailed quantitative study of the expression of the AS-C receptor genes in an adult insect. The tissue distribution of the two AS-C receptors differed significantly; the mRNA levels of AeAS-CrB in the Malpighian tubules were the highest detected, while transcripts for AeAS-CrA were relatively low in this tissue. In addition, the transcript levels of both receptors were different in the thoracic and abdominal ganglia, corpora allata (CA) and the testis of the male. In the CA, the AeAS-CrB mRNA levels were constant from 0 to 72 h after female emergence, while the AeAS-CrA levels increased at 72 h. To complement the receptor expression studies, we analyzed the tissue specificity for allatostatin-C mRNA in female mosquitoes. Expression was high in abdominal ganglia and brain. Transcript levels of allatostatin-C in the head of females were elevated at eclosion and there were no major changes during the first week of adult life or after blood feeding. Fluorometric Imaging Plate Reader (FLIPR) recordings of calcium transients in HEK293T cells transiently expressing both putative receptors showed that they both responded selectively to allatostatin-C stimulation in the nanomolar concentration range. However, the peptide showed slightly greater affinity for AeAS-CrB than AeAS-CrA. Our studies suggest that some of the pleiotropic effects of allatostatin-C in mosquitoes could be mediated by the different receptor paralogs. Transcriptional regulation of the AS-C receptors may not have a critical role in the changes of CA responsiveness to the peptide that we previously described.


Asunto(s)
Culicidae/metabolismo , Neuropéptidos/metabolismo , Receptores de Neuropéptido/metabolismo , Animales , Línea Celular , Humanos , Inmunohistoquímica , Hormonas Juveniles/metabolismo , Filogenia , Reacción en Cadena de la Polimerasa , Receptores de Neuropéptido/clasificación , Receptores de Neuropéptido/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA