Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Med Internet Res ; 25: e44764, 2023 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-37874638

RESUMEN

BACKGROUND: Digital therapeutics (DTx) are software-based products that prevent, manage, or treat a medical condition and are delivered through a smartphone app, web application, or wearable device. Clinical trials assessing DTx pose challenges, foremost among which is designing appropriate digital shams (or digital placebos), which should ideally mimic DTx (in terms of design, components, and duration of treatment) while omitting the active principle or component. OBJECTIVE: The objective of our review was to understand how digital shams are being used in clinical research on DTx in neuroscience, which is the most common therapy area for DTx. METHODS: We conducted a systematic literature review of DTx in neuroscience (including neurodevelopmental, neurodegenerative, and psychiatric disorders) with a focus on controlled clinical trials involving digital shams. Studies were identified from trial registries (ClinicalTrials.gov, the European Union Clinical Trials Register, and Trial Trove) and through structured searches in MEDLINE and Embase (both via the Embase website) and were limited to articles in English published from 2010 onward. These were supplemented by keyword-based searches in PubMed, Google, and Google Scholar and bibliographic searches. Studies assessing DTx in neuroscience (including neurodevelopmental, neurodegenerative, and psychiatric disorders) were included. Details related to the publication, DTx, comparator, patient population, and outcomes were extracted and analyzed. RESULTS: Our search criteria identified 461 neuroscience studies involving 213 unique DTx. Most DTx were extended reality based (86/213, 40.4%) or mobile device based (56/213, 26.3%); 313 were comparative, of which 68 (21.7%) used shams. The most common therapeutic areas assessed in these studies were stroke (42/213, 19.7%), depression (32/213, 15%), and anxiety (24/213, 11.3%). The most common treatments were cognitive behavioral therapy or behavioral therapy (67/213, 32.4%), physical rehabilitation (60/213, 28.2%), and cognitive training (41/213, 19.2%). We identified the following important issues related to the use of digital shams in neuroscience: shams were not validated before use in studies, they varied widely in design (from being nearly identical to the DTx to using different software programs altogether), and the level of patient engagement or satisfaction with the sham and the impact of the sham on study outcomes were infrequently reported. CONCLUSIONS: Digital shams are critical for the clinical development of DTx in neuroscience. Given the importance of sham controls in evaluating DTx efficacy, we provide recommendations on the key information that should be reported in a well-designed DTx trial and propose an algorithm to allow the correct interpretation of DTx study results. Sham-controlled studies should be routinely used in DTx trials-in early-phase studies-to help identify DTx active components and-in late-phase studies-to confirm the efficacy of DTx. The use of shams early in development will ensure that the appropriate sham control is used in later confirmatory trials.


Asunto(s)
Trastornos Mentales , Aplicaciones Móviles , Humanos , Computadoras de Mano , Ansiedad
2.
Mol Ther ; 27(10): 1836-1847, 2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31331814

RESUMEN

CLN6-Batten disease, a form of neuronal ceroid lipofuscinosis is a rare lysosomal storage disorder presenting with gradual declines in motor, visual, and cognitive abilities and early death by 12-15 years of age. We developed a self-complementary adeno-associated virus serotype 9 (scAAV9) vector expressing the human CLN6 gene under the control of a chicken ß-actin (CB) hybrid promoter. Intrathecal delivery of scAAV9.CB.hCLN6 into the cerebrospinal fluid (CSF) of the lumbar spinal cord of 4-year-old non-human primates was safe, well tolerated, and led to efficient targeting throughout the brain and spinal cord. A single intracerebroventricular (i.c.v.) injection at post-natal day 1 in Cln6 mutant mice delivered scAAV9.CB.CLN6 directly into the CSF, and it prevented or drastically reduced all of the pathological hallmarks of Batten disease. Moreover, there were significant improvements in motor performance, learning and memory deficits, and survival in treated Cln6 mutant mice, extending survival from 15 months of age (untreated) to beyond 21 months of age (treated). Additionally, many parameters were similar to wild-type counterparts throughout the lifespan of the treated mice.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Proteínas de la Membrana/genética , Lipofuscinosis Ceroideas Neuronales/psicología , Lipofuscinosis Ceroideas Neuronales/terapia , Actinas/genética , Animales , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Infusiones Intraventriculares , Inyecciones Espinales , Aprendizaje/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Actividad Motora/efectos de los fármacos , Mutación , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo , Primates , Regiones Promotoras Genéticas , Resultado del Tratamiento
3.
Mol Cell Neurosci ; 80: 180-191, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27965018

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating disease characterized by the progressive loss of motor neurons. Neurons, astrocytes, oligodendrocytes and microglial cells all undergo pathological modifications in the onset and progression of ALS. A number of genes involved in the etiopathology of the disease have been identified, but a complete understanding of the molecular mechanisms of ALS has yet to be determined. Currently, people affected by ALS have a life expectancy of only two to five years from diagnosis. The search for a treatment has been slow and mostly unsuccessful, leaving patients in desperate need of better therapies. Until recently, most pre-clinical studies utilized the available ALS animal models. In the past years, the development of new protocols for isolation of patient cells and differentiation into relevant cell types has provided new tools to model ALS, potentially more relevant to the disease itself as they directly come from patients. The use of stem cells is showing promise to facilitate ALS research by expanding our understanding of the disease and help to identify potential new therapeutic targets and therapies to help patients. Advancements in high content analysis (HCA) have the power to contribute to move ALS research forward by combining automated image acquisition along with digital image analysis. With modern HCA machines it is possible, in a period of just a few hours, to observe changes in morphology and survival of cells, under the stimulation of hundreds, if not thousands of drugs and compounds. In this article, we will summarize the major molecular and cellular hallmarks of ALS, describe the advancements provided by the in vitro models developed in the last few years, and review the studies that have applied HCA to the ALS field to date.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Humanos
4.
J Neurosci ; 36(18): 5128-43, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27147664

RESUMEN

UNLABELLED: Tumor necrosis factor (TNF) is associated with the pathophysiology of various neurological disorders, including multiple sclerosis. It exists as a transmembrane form tmTNF, signaling via TNF receptor 2 (TNFR2) and TNFR1, and a soluble form, solTNF, signaling via TNFR1. Multiple sclerosis is associated with the detrimental effects of solTNF acting through TNFR1, while tmTNF promotes repair and remyelination. Here we demonstrate that oligodendroglial TNFR2 is a key mediator of tmTNF-dependent protection in experimental autoimmune encephalomyelitis (EAE). CNP-cre:TNFR2(fl/fl) mice with TNFR2 ablation in oligodendrocytes show exacerbation of the disease with increased axon and myelin pathology, reduced remyelination, and increased loss of oligodendrocyte precursor cells and mature oligodendrocytes. The clinical course of EAE is not improved by the solTNF inhibitor XPro1595 in CNP-cre:TNFR2(fl/fl) mice, indicating that for tmTNF to promote recovery TNFR2 in oligodendrocytes is required. We show that TNFR2 drives differentiation of oligodendrocyte precursor cells, but not proliferation or survival. TNFR2 ablation leads to dysregulated expression of microRNAs, among which are regulators of oligodendrocyte differentiation and inflammation, including miR-7a. Our data provide the first direct in vivo evidence that TNFR2 in oligodendrocytes is important for oligodendrocyte differentiation, thereby sustaining tmTNF-dependent repair in neuroimmune disease. Our studies identify TNFR2 in the CNS as a molecular target for the development of remyelinating agents, addressing the most pressing need in multiple sclerosis therapy nowadays. SIGNIFICANCE STATEMENT: Our study, using novel TNF receptor 2 (TNFR2) conditional KO mice with selective TNFR2 ablation in oligodendrocytes, provides the first direct evidence that TNFR2 is an important signal for oligodendrocyte differentiation. Following activation by transmembrane TNF, TNFR2 initiates pathways that drive oligodendrocytes into a reparative mode contributing to remyelination following disease. This identifies TNFR2 as a new molecular target for the development of therapeutic agents in multiple sclerosis.


Asunto(s)
Diferenciación Celular/genética , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Vaina de Mielina , Neuroglía/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Animales , Axones/patología , Conducta Animal , Supervivencia Celular/genética , Encefalomielitis Autoinmune Experimental/psicología , Femenino , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Noqueados , Regeneración Nerviosa/genética , Células-Madre Neurales , Factor de Necrosis Tumoral alfa/metabolismo
5.
Proc Natl Acad Sci U S A ; 109(19): 7517-22, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22529377

RESUMEN

Axon regeneration in the central nervous system normally fails, in part because of a developmental decline in the intrinsic ability of CNS projection neurons to extend axons. Members of the KLF family of transcription factors regulate regenerative potential in developing CNS neurons. Expression of one family member, KLF7, is down-regulated developmentally, and overexpression of KLF7 in cortical neurons in vitro promotes axonal growth. To circumvent difficulties in achieving high neuronal expression of exogenous KLF7, we created a chimera with the VP16 transactivation domain, which displayed enhanced neuronal expression compared with the native protein while maintaining transcriptional activation and growth promotion in vitro. Overexpression of VP16-KLF7 overcame the developmental loss of regenerative ability in cortical slice cultures. Adult corticospinal tract (CST) neurons failed to up-regulate KLF7 in response to axon injury, and overexpression of VP16-KLF7 in vivo promoted both sprouting and regenerative axon growth in the CST of adult mice. These findings identify a unique means of promoting CST axon regeneration in vivo by reengineering a developmentally down-regulated, growth-promoting transcription factor.


Asunto(s)
Axones/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Regeneración Nerviosa/fisiología , Tractos Piramidales/fisiología , Animales , Axones/metabolismo , Células Cultivadas , Etopósido , Femenino , Expresión Génica , Ingeniería Genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Proteína Vmw65 de Virus del Herpes Simple/genética , Humanos , Inmunohistoquímica , Factores de Transcripción de Tipo Kruppel/genética , Mediciones Luminiscentes/métodos , Ratones , Ratones Endogámicos C57BL , Mutación , Regeneración Nerviosa/genética , Neuritas/metabolismo , Neuritas/fisiología , Neuronas/citología , Neuronas/metabolismo , Neuronas/fisiología , Tractos Piramidales/citología , Tractos Piramidales/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Activación Transcripcional
6.
Clin Pharmacol Ther ; 114(3): 578-590, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37392464

RESUMEN

The promise of transforming digital technologies into treatments is what drives the development of digital therapeutics (DTx), generally known as software applications embedded within accessible technologies-such as smartphones-to treat, manage, or prevent a pathological condition. Whereas DTx solutions that successfully demonstrate effectiveness and safety could drastically improve the life of patients in multiple therapeutic areas, there is a general consensus that generating therapeutic evidence for DTx presents challenges and open questions. We believe there are three main areas where the application of clinical pharmacology principles from the drug development field could benefit DTx development: the characterization of the mechanism of action, the optimization of the intervention, and, finally, its dosing. We reviewed DTx studies to explore how the field is approaching these topics and to better characterize the challenges associated with them. This leads us to emphasize the role that the application of clinical pharmacology principles could play in the development of DTx and to advocate for a development approach that merges such principles from development of traditional therapeutics with important considerations from the highly attractive and fast-paced world of digital solutions.


Asunto(s)
Farmacología Clínica , Programas Informáticos , Terapéutica , Humanos
7.
J Neurochem ; 122(3): 568-81, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22640015

RESUMEN

The R7 family of regulators of G protein signaling (RGS) is involved in many functions of the nervous system. This family includes RGS6, RGS7, RGS9, and RGS11 gene products and is defined by the presence of the characteristic first found in Disheveled, Egl-10, Pleckstrin (DEP), DEP helical extension (DHEX), Gγ-like, and RGS domains. Herein, we examined the subcellular localization of RGS7, the most broadly expressed R7 member. Our immunofluorescence studies of retinal and dorsal root ganglion neurons showed that RGS7 concentrated at the plasma membrane of cell bodies, in structures resembling lamellipodia or filopodia along the processes, and at the dendritic tips. At the plasma membrane of dorsal root ganglia neurons, RGS7 co-localized with its known binding partners R7 RGS binding protein (R7BP), Gαo, and Gαq. More than 50% of total RGS7-specific immunofluorescence was present in the cytoplasm, primarily within numerous small puncta that did not co-localize with R7BP. No specific RGS7 or R7BP immunoreactivity was detected in the nuclei. In transfected cell lines, ectopic RGS7 had both diffuse cytosolic and punctate localization patterns. RGS7 also localized in centrosomes. Structure-function analysis showed that the punctate localization was mediated by the DEP/DHEX domains, and centrosomal localization was dependent on the DHEX domain.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Neuronas/metabolismo , Proteínas RGS/metabolismo , Fracciones Subcelulares/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cricetinae , Cricetulus , Subunidades beta de la Proteína de Unión al GTP/deficiencia , Ganglios Espinales/citología , Regulación de la Expresión Génica/genética , Imagenología Tridimensional , Inmunoprecipitación , Técnicas In Vitro , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Mutación/genética , Neuronas/citología , Conformación Proteica , Proteínas RGS/genética , Retina/citología , Retina/metabolismo , Transfección
8.
Eur J Neurosci ; 32(8): 1364-79, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20950280

RESUMEN

We report gene profiling data on genomic processes underlying the progression towards recurrent seizures after injection of kainic acid (KA) into the mouse hippocampus. Focal injection enabled us to separate the effects of proepileptic stimuli initiated by KA injection. Both the injected and contralateral hippocampus participated in the status epilepticus. However, neuronal death induced by KA treatment was restricted to the injected hippocampus, although there was some contralateral axonal degeneration. We profiled gene expression changes in dorsal and ventral regions of both the injected and contralateral hippocampus. Changes were detected in the expression of 1526 transcripts in samples from three time-points: (i) during the KA-induced status epilepticus, (ii) at 2 weeks, before recurrent seizures emerged, and (iii) at 6 months after seizures emerged. Grouping genes with similar spatio-temporal changes revealed an early transcriptional response, strong immune, cell death and growth responses at 2 weeks and an activation of immune and extracellular matrix genes persisting at 6 months. Immunostaining for proteins coded by genes identified from array studies provided evidence for gliogenesis and suggested that the proteoglycan biglycan is synthesized by astrocytes and contributes to a glial scar. Gene changes at 6 months after KA injection were largely restricted to tissue from the injection site. This suggests that either recurrent seizures might depend on maintained processes including immune responses and changes in extracellular matrix proteins near the injection site or alternatively might result from processes, such as growth, distant from the injection site and terminated while seizures are maintained.


Asunto(s)
Expresión Génica , Hipocampo/fisiopatología , Neuronas/fisiología , Convulsiones/genética , Convulsiones/fisiopatología , Animales , Muerte Celular , Hipocampo/metabolismo , Inmunohistoquímica , Ácido Kaínico/administración & dosificación , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Convulsiones/inducido químicamente , Convulsiones/metabolismo
9.
Methods Mol Biol ; 1683: 293-304, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29082499

RESUMEN

High Content Screening (HCS) can be used to analyze the morphology of neuronal primary cultures on a large scale. When used in the field of neuronal regeneration this approach allows the screening of hundreds or thousands of perturbagens, such as miRNAs, cDNAs, or compounds, for their ability to induce neuronal growth. One of the most important steps while designing these kinds of experiments is the choice of the correct neuronal model. Testing the correct neuronal type is critical to obtain results that are biologically significant and that can later be translated to a clinical setting. For example, if the goal is identifying possible therapies for Spinal Cord Injury (SCI), a challenging target is the neuronal projection from the motor cortex to the spinal cord, the corticospinal tract. Here, we describe the experimental protocols that can be used to produce primary cortical culture from young rat cortices, electroporate the neurons to study the effect of altered gene expression on neurite growth, and immunostain to measure neurite growth parameters.


Asunto(s)
Corteza Cerebral/citología , Ensayos Analíticos de Alto Rendimiento , Neuronas/fisiología , Fenotipo , Animales , Descubrimiento de Drogas , Regulación de la Expresión Génica/efectos de los fármacos , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/genética , Neuronas/efectos de los fármacos , Ratas
10.
FEBS Lett ; 591(14): 2091-2105, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28626869

RESUMEN

Peripheral neurons regenerate their axons after injury. Transcriptional regulation by microRNAs (miRNAs) is one possible mechanism controlling regeneration. We profiled miRNA expression in mouse dorsal root ganglion neurons after a sciatic nerve crush, and identified 49 differentially expressed miRNAs. We evaluated the functional role of each miRNA using a phenotypic analysis approach. To predict the targets of the miRNAs we employed RNA-Sequencing and examined transcription at the isoform level. We identify thousands of differentially expressed isoforms and bioinformatically associate the miRNAs that modulate neurite growth with their putative target isoforms to outline a network of regulatory events underlying peripheral nerve regeneration. MiR-298, let-7a, and let-7f enhance neurite growth and target the majority of isoforms in the differentially expressed network.


Asunto(s)
Ganglios Espinales/citología , MicroARNs/genética , Proyección Neuronal/genética , Animales , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Fenotipo , Isoformas de ARN/genética , Análisis de Secuencia de ARN , Transcripción Genética
11.
eNeuro ; 4(4)2017.
Artículo en Inglés | MEDLINE | ID: mdl-28828403

RESUMEN

Stress and glucocorticoid (GC) release are common behavioral and hormonal responses to injury or disease. In the brain, stress/GCs can alter neuron structure and function leading to cognitive impairment. Stress and GCs also exacerbate pain, but whether a corresponding change occurs in structural plasticity of sensory neurons is unknown. Here, we show that in female mice (Mus musculus) basal GC receptor (Nr3c1, also known as GR) expression in dorsal root ganglion (DRG) sensory neurons is 15-fold higher than in neurons in canonical stress-responsive brain regions (M. musculus). In response to stress or GCs, adult DRG neurite growth increases through mechanisms involving GR-dependent gene transcription. In vivo, prior exposure to an acute systemic stress increases peripheral nerve regeneration. These data have broad clinical implications and highlight the importance of stress and GCs as novel behavioral and circulating modifiers of neuronal plasticity.


Asunto(s)
Axones/fisiología , Regeneración Nerviosa/fisiología , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/complicaciones , Estrés Psicológico/patología , Activación Transcripcional/fisiología , Factor de Transcripción Activador 3/metabolismo , Animales , Proteínas de Unión al Calcio , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Antagonistas de Hormonas/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mifepristona/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuritas/patología , Neuropatía Ciática/fisiopatología , Células Receptoras Sensoriales/fisiología , Estatmina
12.
FEBS Lett ; 580(18): 4449-56, 2006 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-16870183

RESUMEN

We have studied the splicing regulation of NF1 exons 36 and 37. We show that they not only require an intact exonic Splicing Enhancer (ESE) within exon 37, but also need the genomic region stretching from exons 31 to 38. Any nucleotide change in two exon 37 third codon positions disrupts the ESE. The extent of exons 36 and 37 skipping due to a mutated ESE depends on the genomic context. This is a unique example of what may be a more general phenomena involved in the tuning of pre-mRNA processing and gene expression modulation in the chromosomal setting.


Asunto(s)
Empalme Alternativo , Neurofibromatosis 1/genética , Secuencias Reguladoras de Ácido Ribonucleico , Exones , Genómica , Células HeLa , Humanos , Neurofibromatosis 1/metabolismo , Mutación Puntual , Sitios de Empalme de ARN , ARN Mensajero/metabolismo
13.
Neural Regen Res ; 11(1): 144-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26981104

RESUMEN

Animal models of sciatic nerve injury are commonly used to study neuropathic pain as well as axon regeneration. Administration of post-surgical analgesics is an important consideration for animal welfare, but the actions of the analgesic must not interfere with the scientific goals of the experiment. In this study, we show that treatment with either buprenorphine or acetaminophen following a bilateral sciatic nerve crush surgery does not alter the expression in dorsal root ganglion (DRG) sensory neurons of a panel of genes associated with wound healing. These findings indicate that the post-operative use of buprenorphine or acetaminophen at doses commonly suggested by Institutional Animal Care and Use Committees does not change the intrinsic gene expression response of DRG neurons to a sciatic nerve crush injury, for many wound healing-associated genes. Therefore, administration of post-operative analgesics may not confound the results of transcriptomic studies employing this injury model.

14.
Front Genet ; 6: 263, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26322075

RESUMEN

CGG repeat expansions in the Fragile X mental retardation 1 (FMR1) gene are responsible for a family of associated disorders characterized by either intellectual disability and autism Fragile X Syndrome (FXS), or adult-onset neurodegeneration Fragile X-associated Tremor/Ataxia Syndrome. However, the FMR1 locus is complex and encodes several long non-coding RNAs, whose expression is altered by repeat expansion mutations. The role of these lncRNAs is thus far unknown; therefore we investigated the functionality of FMR4, which we previously identified. "Full"-length expansions of the FMR1 triplet repeat cause silencing of both FMR1 and FMR4, thus we are interested in potential loss-of-function that may add to phenotypic manifestation of FXS. Since the two transcripts do not exhibit cis-regulation of one another, we examined the potential for FMR4 to regulate target genes at distal genomic loci using gene expression microarrays. We identified FMR4-responsive genes, including the methyl-CpG-binding domain protein 4 (MBD4). Furthermore, we found that in differentiating human neural precursor cells, FMR4 expression is developmentally regulated in opposition to expression of both FMR1 (which is expected to share a bidirectional promoter with FMR4) and MBD4. We therefore propose that FMR4's function is as a gene-regulatory lncRNA and that this transcript may function in normal development. Closer examination of FMR4 increases our understanding of the role of regulatory lncRNA and the consequences of FMR1 repeat expansions.

15.
Brain Res ; 1533: 122-30, 2013 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-23948100

RESUMEN

Previously we reported that several microRNAs (miRNA) are upregulated following experimentally induced traumatic brain injury (TBI) using both in vivo and in vitro approaches. Specific miRNAs were found to be sensitive to therapeutic hypothermia and may therefore be important targets for neuroprotective strategies. In this study we developed plasmid constructs that overexpress temperature sensitive miRNAs: miR-34a, miR-451, and miR-874. These constructs were transfected into cultured cortical neurons that were subjected to stretch injury using a cell injury controller device. Levels of expression of genes associated with stress, inflammation, apoptosis and transcriptional regulation were measured by qRT-PCR. mRNA levels of cytokines interleukin 1-ß (IL1-ß) and tumor necrosis factor alpha (TNF-α) as well as heat shock protein 70 (HSP70) and Caspase 11 were found to be increased up to 24 fold higher than controls in cells overexpressing these miRNAs. After moderate stretch injury, the expression of IL1-ß, TNF-α, HSP70 and Caspase 11 all increased over control levels found in uninjured cells suggesting that overexpression of these miRNAs increases cellular vulnerability. miR-34a directly inhibits Bcl2 and XIAP, both anti-apoptotic proteins. The observed increase in Caspase 11 with over-expression of miR-34a indicates that miR-34a may be inducing apoptosis by reducing the levels of anti-apoptotic proteins. miR-34a is predicted to inhibit Jun, which was seen to decrease in cells overexpressing this miRNA along with Fos. Over expression of several miRNAs found to be induced by TBI in vivo (miR-34a, miR-451 and miR-874) leads to increased vulnerability in transfected neurons. Therapeutic hypothermia blunts the expression of these miRNAs in vivo and antisense silencing could be a potential therapeutic approach to targeting the consequences of TBI.


Asunto(s)
Corteza Cerebral/metabolismo , MicroARNs/metabolismo , Neuronas/metabolismo , Animales , Apoptosis/genética , Lesiones Encefálicas/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Ratas
16.
Semin Fetal Neonatal Med ; 17(6): 347-52, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22906916

RESUMEN

The importance of the involvement of non-protein coding RNAs in biological processes has become evident in recent years along with the identification of the transcriptional regulatory mechanisms that allow them to exert their roles. MicroRNAs (miRNAs) are a novel class of small non-coding RNA that regulates messenger RNA abundance. The capacity of each miRNA to target several transcripts suggests an ability to build a complex regulatory network for fine tuning gene expression; a mechanism by which they are thought to regulate cell fate, proliferation and identity. The brain expresses more distinct miRNAs than any other tissue in vertebrates and it presents an impressive variety of cell types, including many different classes of neurons. Here we review more than 10 years of miRNA research, and discuss the most important findings that have established miRNAs as key regulators of neuronal development.


Asunto(s)
Encéfalo , Redes Reguladoras de Genes/fisiología , MicroARNs/genética , Neurogénesis/genética , Biosíntesis de Proteínas , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Fenómenos Fisiológicos Celulares/genética , Regulación de la Expresión Génica , Código Genético , Investigación Genética , Humanos , ARN Pequeño no Traducido/genética
17.
PLoS One ; 7(1): e30417, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22272348

RESUMEN

To fully understand cell type identity and function in the nervous system there is a need to understand neuronal gene expression at the level of isoform diversity. Here we applied Next Generation Sequencing of the transcriptome (RNA-Seq) to purified sensory neurons and cerebellar granular neurons (CGNs) grown on an axonal growth permissive substrate. The goal of the analysis was to uncover neuronal type specific isoforms as a prelude to understanding patterns of gene expression underlying their intrinsic growth abilities. Global gene expression patterns were comparable to those found for other cell types, in that a vast majority of genes were expressed at low abundance. Nearly 18% of gene loci produced more than one transcript. More than 8000 isoforms were differentially expressed, either to different degrees in different neuronal types or uniquely expressed in one or the other. Sensory neurons expressed a larger number of genes and gene isoforms than did CGNs. To begin to understand the mechanisms responsible for the differential gene/isoform expression we identified transcription factor binding sites present specifically in the upstream genomic sequences of differentially expressed isoforms, and analyzed the 3' untranslated regions (3' UTRs) for microRNA (miRNA) target sites. Our analysis defines isoform diversity for two neuronal types with diverse axon growth capabilities and begins to elucidate the complex transcriptional landscape in two neuronal populations.


Asunto(s)
Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neuronas/metabolismo , Células Receptoras Sensoriales/metabolismo , Transcriptoma , Regiones no Traducidas 5'/genética , Animales , Sitios de Unión/genética , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Cerebelo/citología , Cerebelo/metabolismo , Análisis por Conglomerados , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Modelos Logísticos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Sistemas de Lectura Abierta/genética , Sistema Nervioso Periférico/citología , Sistema Nervioso Periférico/metabolismo , Isoformas de Proteínas/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción
18.
Genome Res ; 19(2): 255-65, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074369

RESUMEN

Finding and characterizing mRNAs, their transcription start sites (TSS), and their associated promoters is a major focus in post-genome biology. Mammalian cells have at least 5-10 magnitudes more TSS than previously believed, and deeper sequencing is necessary to detect all active promoters in a given tissue. Here, we present a new method for high-throughput sequencing of 5' cDNA tags-DeepCAGE: merging the Cap Analysis of Gene Expression method with ultra-high-throughput sequence technology. We apply DeepCAGE to characterize 1.4 million sequenced TSS from mouse hippocampus and reveal a wealth of novel core promoters that are preferentially used in hippocampus: This is the most comprehensive promoter data set for any tissue to date. Using these data, we present evidence indicating a key role for the Arnt2 transcription factor in hippocampus gene regulation. DeepCAGE can also detect promoters used only in a small subset of cells within the complex tissue.


Asunto(s)
Hipocampo/metabolismo , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN/métodos , Animales , Sitios de Unión , Mapeo Cromosómico/métodos , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Especificidad de Órganos/genética , Unión Proteica , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA