Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 83(13): 2258-2275.e11, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-37369199

RESUMEN

The pre-mRNA life cycle requires intron processing; yet, how intron-processing defects influence splicing and gene expression is unclear. Here, we find that TTDN1/MPLKIP, which is encoded by a gene implicated in non-photosensitive trichothiodystrophy (NP-TTD), functionally links intron lariat processing to spliceosomal function. The conserved TTDN1 C-terminal region directly binds lariat debranching enzyme DBR1, whereas its N-terminal intrinsically disordered region (IDR) binds the intron-binding complex (IBC). TTDN1 loss, or a mutated IDR, causes significant intron lariat accumulation, as well as splicing and gene expression defects, mirroring phenotypes observed in NP-TTD patient cells. A Ttdn1-deficient mouse model recapitulates intron-processing defects and certain neurodevelopmental phenotypes seen in NP-TTD. Fusing DBR1 to the TTDN1 IDR is sufficient to recruit DBR1 to the IBC and circumvents the functional requirement for TTDN1. Collectively, our findings link RNA lariat processing with splicing outcomes by revealing the molecular function of TTDN1.


Asunto(s)
Síndromes de Tricotiodistrofia , Animales , Ratones , Intrones/genética , Síndromes de Tricotiodistrofia/genética , ARN Nucleotidiltransferasas/genética , Empalme del ARN
2.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34732576

RESUMEN

ATP-sensitive potassium (KATP) gain-of-function (GOF) mutations cause neonatal diabetes, with some individuals exhibiting developmental delay, epilepsy, and neonatal diabetes (DEND) syndrome. Mice expressing KATP-GOF mutations pan-neuronally (nKATP-GOF) demonstrated sensorimotor and cognitive deficits, whereas hippocampus-specific hKATP-GOF mice exhibited mostly learning and memory deficiencies. Both nKATP-GOF and hKATP-GOF mice showed altered neuronal excitability and reduced hippocampal long-term potentiation (LTP). Sulfonylurea therapy, which inhibits KATP, mildly improved sensorimotor but not cognitive deficits in KATP-GOF mice. Mice expressing KATP-GOF mutations in pancreatic ß-cells developed severe diabetes but did not show learning and memory deficits, suggesting neuronal KATP-GOF as promoting these features. These findings suggest a possible origin of cognitive dysfunction in DEND and the need for novel drugs to treat neurological features induced by neuronal KATP-GOF.


Asunto(s)
Trastornos del Conocimiento/etiología , Diabetes Mellitus/psicología , Epilepsia/psicología , Hipocampo/metabolismo , Enfermedades del Recién Nacido/psicología , Canales KATP/genética , Trastornos Motores/etiología , Trastornos Psicomotores/psicología , Animales , Diabetes Mellitus/etiología , Diabetes Mellitus/metabolismo , Modelos Animales de Enfermedad , Epilepsia/etiología , Epilepsia/metabolismo , Femenino , Mutación con Ganancia de Función , Enfermedades del Recién Nacido/etiología , Enfermedades del Recién Nacido/metabolismo , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/etiología , Potenciación a Largo Plazo , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Ratones Transgénicos , Trastornos Psicomotores/etiología , Trastornos Psicomotores/metabolismo , Compuestos de Sulfonilurea/uso terapéutico
3.
Proc Natl Acad Sci U S A ; 116(47): 23822-23828, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31694884

RESUMEN

Nicotinamide adenine dinucleotide (NAD+) is a critical coenzyme for cellular energy metabolism. The aim of the present study was to determine the importance of brown and white adipose tissue (BAT and WAT) NAD+ metabolism in regulating whole-body thermogenesis and energy metabolism. Accordingly, we generated and analyzed adipocyte-specific nicotinamide phosphoribosyltransferase (Nampt) knockout (ANKO) and brown adipocyte-specific Nampt knockout (BANKO) mice because NAMPT is the rate-limiting NAD+ biosynthetic enzyme. We found ANKO mice, which lack NAMPT in both BAT and WAT, had impaired gene programs involved in thermogenesis and mitochondrial function in BAT and a blunted thermogenic (rectal temperature, BAT temperature, and whole-body oxygen consumption) response to acute cold exposure, prolonged fasting, and administration of ß-adrenergic agonists (norepinephrine and CL-316243). In addition, the absence of NAMPT in WAT markedly reduced adrenergic-mediated lipolytic activity, likely through inactivation of the NAD+-SIRT1-caveolin-1 axis, which limits an important fuel source fatty acid for BAT thermogenesis. These metabolic abnormalities were rescued by treatment with nicotinamide mononucleotide (NMN), which bypasses the block in NAD+ synthesis induced by NAMPT deficiency. Although BANKO mice, which lack NAMPT in BAT only, had BAT cellular alterations similar to the ANKO mice, BANKO mice had normal thermogenic and lipolytic responses. We also found NAMPT expression in supraclavicular adipose tissue (where human BAT is localized) obtained from human subjects increased during cold exposure, suggesting our finding in rodents could apply to people. These results demonstrate that adipose NAMPT-mediated NAD+ biosynthesis is essential for regulating adaptive thermogenesis, lipolysis, and whole-body energy metabolism.


Asunto(s)
Adaptación Fisiológica , Tejido Adiposo Pardo/metabolismo , Metabolismo Energético , Homeostasis , NAD/biosíntesis , Termogénesis , Tejido Adiposo Pardo/enzimología , Animales , Caveolina 1/antagonistas & inhibidores , Frío , Citocinas/genética , Ayuno , Humanos , Ratones , Ratones Noqueados , Mononucleótido de Nicotinamida/administración & dosificación , Nicotinamida Fosforribosiltransferasa/genética
4.
Proc Natl Acad Sci U S A ; 116(40): 20097-20103, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31527255

RESUMEN

Infantile globoid cell leukodystrophy (GLD, Krabbe disease) is a fatal demyelinating disorder caused by a deficiency in the lysosomal enzyme galactosylceramidase (GALC). GALC deficiency leads to the accumulation of the cytotoxic glycolipid, galactosylsphingosine (psychosine). Complementary evidence suggested that psychosine is synthesized via an anabolic pathway. Here, we show instead that psychosine is generated catabolically through the deacylation of galactosylceramide by acid ceramidase (ACDase). This reaction uncouples GALC deficiency from psychosine accumulation, allowing us to test the long-standing "psychosine hypothesis." We demonstrate that genetic loss of ACDase activity (Farber disease) in the GALC-deficient mouse model of human GLD (twitcher) eliminates psychosine accumulation and cures GLD. These data suggest that ACDase could be a target for substrate reduction therapy (SRT) in Krabbe patients. We show that pharmacological inhibition of ACDase activity with carmofur significantly decreases psychosine accumulation in cells from a Krabbe patient and prolongs the life span of the twitcher (Twi) mouse. Previous SRT experiments in the Twi mouse utilized l-cycloserine, which inhibits an enzyme several steps upstream of psychosine synthesis, thus altering the balance of other important lipids. Drugs that directly inhibit ACDase may have a more acceptable safety profile due to their mechanistic proximity to psychosine biogenesis. In total, these data clarify our understanding of psychosine synthesis, confirm the long-held psychosine hypothesis, and provide the impetus to discover safe and effective inhibitors of ACDase to treat Krabbe disease.


Asunto(s)
Ceramidasa Ácida/genética , Eliminación de Gen , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/metabolismo , Psicosina/metabolismo , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Metilación de ADN , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Leucodistrofia de Células Globoides/tratamiento farmacológico
5.
J Lipid Res ; 62: 100079, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33894211

RESUMEN

Vascular disease contributes to neurodegeneration, which is associated with decreased blood pressure in older humans. Plasmalogens, ether phospholipids produced by peroxisomes, are decreased in Alzheimer's disease, Parkinson's disease, and other neurodegenerative disorders. However, the mechanistic links between ether phospholipids, blood pressure, and neurodegeneration are not fully understood. Here, we show that endothelium-derived ether phospholipids affect blood pressure, behavior, and neurodegeneration in mice. In young adult mice, inducible endothelial-specific disruption of PexRAP, a peroxisomal enzyme required for ether lipid synthesis, unexpectedly decreased circulating plasmalogens. PexRAP endothelial knockout (PEKO) mice responded normally to hindlimb ischemia but had lower blood pressure and increased plasma renin activity. In PEKO as compared with control mice, tyrosine hydroxylase was decreased in the locus coeruleus, which maintains blood pressure and arousal. PEKO mice moved less, slept more, and had impaired attention to and recall of environmental events as well as mild spatial memory deficits. In PEKO hippocampus, gliosis was increased, and a plasmalogen associated with memory was decreased. Despite lower blood pressure, PEKO mice had generally normal homotopic functional connectivity by optical neuroimaging of the cerebral cortex. Decreased glycogen synthase kinase-3 phosphorylation, a marker of neurodegeneration, was detected in PEKO cerebral cortex. In a co-culture system, PexRAP knockdown in brain endothelial cells decreased glycogen synthase kinase-3 phosphorylation in co-cultured astrocytes that was rescued by incubation with the ether lipid alkylglycerol. Taken together, our findings suggest that endothelium-derived ether lipids mediate several biological processes and may also confer neuroprotection in mice.


Asunto(s)
Presión Sanguínea
6.
J Neurosci ; 38(1): 200-219, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29133437

RESUMEN

CCCTC-binding factor (CTCF) is an 11 zinc finger DNA-binding domain protein that regulates gene expression by modifying 3D chromatin structure. Human mutations in CTCF cause intellectual disability and autistic features. Knocking out Ctcf in mouse embryonic neurons is lethal by neonatal age, but the effects of CTCF deficiency in postnatal neurons are less well studied. We knocked out Ctcf postnatally in glutamatergic forebrain neurons under the control of Camk2a-Cre. CtcfloxP/loxP;Camk2a-Cre+ (Ctcf CKO) mice of both sexes were viable and exhibited profound deficits in spatial learning/memory, impaired motor coordination, and decreased sociability by 4 months of age. Ctcf CKO mice also had reduced dendritic spine density in the hippocampus and cerebral cortex. Microarray analysis of mRNA from Ctcf CKO mouse hippocampus identified increased transcription of inflammation-related genes linked to microglia. Separate microarray analysis of mRNA isolated specifically from Ctcf CKO mouse hippocampal neurons by ribosomal affinity purification identified upregulation of chemokine signaling genes, suggesting crosstalk between neurons and microglia in Ctcf CKO hippocampus. Finally, we found that microglia in Ctcf CKO mouse hippocampus had abnormal morphology by Sholl analysis and increased immunostaining for CD68, a marker of microglial activation. Our findings confirm that Ctcf KO in postnatal neurons causes a neurobehavioral phenotype in mice and provide novel evidence that CTCF depletion leads to overexpression of inflammation-related genes and microglial dysfunction.SIGNIFICANCE STATEMENT CCCTC-binding factor (CTCF) is a DNA-binding protein that organizes nuclear chromatin topology. Mutations in CTCF cause intellectual disability and autistic features in humans. CTCF deficiency in embryonic neurons is lethal in mice, but mice with postnatal CTCF depletion are less well studied. We find that mice lacking Ctcf in Camk2a-expressing neurons (Ctcf CKO mice) have spatial learning/memory deficits, impaired fine motor skills, subtly altered social interactions, and decreased dendritic spine density. We demonstrate that Ctcf CKO mice overexpress inflammation-related genes in the brain and have microglia with abnormal morphology that label positive for CD68, a marker of microglial activation. Our findings suggest that inflammation and dysfunctional neuron-microglia interactions are factors in the pathology of CTCF deficiency.


Asunto(s)
Factor de Unión a CCCTC/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Inflamación/genética , Inflamación/patología , Microglía/patología , Neuronas/patología , Transcripción Genética/genética , Animales , Electroencefalografía , Femenino , Expresión Génica/genética , Integrasas , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/genética , Trastornos de la Memoria/psicología , Ratones , Ratones Noqueados , Análisis por Micromatrices , Neuronas/metabolismo , Desempeño Psicomotor , Conducta Social
7.
Neurobiol Learn Mem ; 165: 107087, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31499164

RESUMEN

Intellectual and developmental disabilities (IDDs) are a common group of disorders that frequently share overlapping symptoms, including cognitive deficits, altered attention, seizures, impaired social interactions, and anxiety. The causes of these disorders are varied ranging from early prenatal/postnatal insults to genetic variants that either cause or are associated with an increased likelihood of an IDD. As many of the symptoms observed in individuals with IDDs are a manifestation of altered nervous system function resulting in altered behaviors, it should not be surprising that the field is very dependent upon in vivo model systems. This special issue of Neurobiology of Learning and Memory is focused on the methods and approaches that are being used to model and understand these disorders in mammals. While surveys by the Pew Foundation continue to find a high degree of confidence/trust in scientists by the public, several recent studies have documented issues with reproducibility in scientific publications. This special issue includes both primary research articles and review articles in which careful attention has been made to transparently report methods and use rigorous approaches to ensure reproducibility. Although there have been and will continue to be remarkable advances for treatment of subset of IDDs, it is clear that this field is still in its early stages. There is no doubt that the strategies being used to model IDDs will continue to evolve. We hope this special issue will support this evolution so that we can maintain the trust of the public and elected officials, and continue developing evidence-based approaches to new therapeutics.


Asunto(s)
Discapacidades del Desarrollo/psicología , Modelos Animales de Enfermedad , Discapacidad Intelectual/psicología , Animales , Discapacidades del Desarrollo/etiología , Humanos , Discapacidad Intelectual/etiología
8.
Neurobiol Learn Mem ; 165: 106834, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29550366

RESUMEN

Fifteen years ago Olney and colleagues began using animal models to evaluate the effects of anesthetic and sedative agents (ASAs) on neurodevelopment. The results from ongoing studies indicate that, under certain conditions, exposure to these drugs during development induces an acute elevated apoptotic neurodegenerative response in the brain and long-term functional impairments. These animal models have played a significant role in bringing attention to the possible adverse effects of exposing the developing brain to ASAs when few concerns had been raised previously in the medical community. The apoptotic degenerative response resulting from neonatal exposure to ASAs has been replicated in many studies in both rodents and non-human primates, suggesting that a similar effect may occur in humans. In both rodents and non-human primates, significantly increased levels of apoptotic degeneration are often associated with functional impairments later in life. However, behavioral deficits following developmental ASA exposure have not been consistently reported even when significantly elevated levels of apoptotic degeneration have been documented in animal models. In the present work, we review this literature and propose a rodent model for assessing potential functional deficits following neonatal ASA exposure with special reference to experimental design and procedural issues. Our intent is to improve test sensitivity and replicability for detecting subtle behavioral effects, and thus enhance the translational significance of ASA models.


Asunto(s)
Anestesia/efectos adversos , Trastornos del Neurodesarrollo/inducido químicamente , Anestésicos/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad
9.
J Neurosci ; 34(17): 5800-15, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24760840

RESUMEN

Nicotinamide adenine dinucleotide (NAD(+)) is an enzyme cofactor or cosubstrate in many essential biological pathways. To date, the primary source of neuronal NAD(+) has been unclear. NAD(+) can be synthesized from several different precursors, among which nicotinamide is the substrate predominantly used in mammals. The rate-limiting step in the NAD(+) biosynthetic pathway from nicotinamide is performed by nicotinamide phosphoribosyltransferase (Nampt). Here, we tested the hypothesis that neurons use intracellular Nampt-mediated NAD(+) biosynthesis by generating and evaluating mice lacking Nampt in forebrain excitatory neurons (CaMKIIαNampt(-/-) mice). CaMKIIαNampt(-/-) mice showed hippocampal and cortical atrophy, astrogliosis, microgliosis, and abnormal CA1 dendritic morphology by 2-3 months of age. Importantly, these histological changes occurred with altered intrahippocampal connectivity and abnormal behavior; including hyperactivity, some defects in motor skills, memory impairment, and reduced anxiety, but in the absence of impaired sensory processes or long-term potentiation of the Schaffer collateral pathway. These results clearly demonstrate that forebrain excitatory neurons mainly use intracellular Nampt-mediated NAD(+) biosynthesis to mediate their survival and function. Studying this particular NAD(+) biosynthetic pathway in these neurons provides critical insight into their vulnerability to pathophysiological stimuli and the development of therapeutic and preventive interventions for their preservation.


Asunto(s)
Corteza Cerebral/metabolismo , Cognición/fisiología , Hipocampo/metabolismo , Neuronas/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Animales , Atrofia/genética , Atrofia/metabolismo , Atrofia/patología , Conducta Animal/fisiología , Corteza Cerebral/patología , Gliosis/metabolismo , Gliosis/patología , Hipocampo/patología , Memoria/fisiología , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Red Nerviosa/metabolismo , Red Nerviosa/patología , Neuronas/patología , Nicotinamida Fosforribosiltransferasa/genética
10.
J Neurosci ; 34(21): 7281-92, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24849360

RESUMEN

Apolipoprotein E (apoE) is the strongest known genetic risk factor for late onset Alzheimer's disease (AD). It influences amyloid-ß (Aß) clearance and aggregation, which likely contributes in large part to its role in AD pathogenesis. We recently found that HJ6.3, a monoclonal antibody against apoE, significantly reduced Aß plaque load when given to APPswe/PS1ΔE9 (APP/PS1) mice starting before the onset of plaque deposition. To determine whether the anti-apoE antibody HJ6.3 affects Aß plaques, neuronal network function, and behavior in APP/PS1 mice after plaque onset, we administered HJ6.3 (10 mg/kg/week) or PBS intraperitoneally to 7-month-old APP/PS1 mice for 21 weeks. HJ6.3 mildly improved spatial learning performance in the water maze, restored resting-state functional connectivity, and modestly reduced brain Aß plaque load. There was no effect of HJ6.3 on total plasma cholesterol or cerebral amyloid angiopathy. To investigate the underlying mechanisms of anti-apoE immunotherapy, HJ6.3 was applied to the brain cortical surface and amyloid deposition was followed over 2 weeks using in vivo imaging. Acute exposure to HJ6.3 affected the course of amyloid deposition in that it prevented the formation of new amyloid deposits, limited their growth, and was associated with occasional clearance of plaques, a process likely associated with direct binding to amyloid aggregates. Topical application of HJ6.3 for only 14 d also decreased the density of amyloid plaques assessed postmortem. Collectively, these studies suggest that anti-apoE antibodies have therapeutic potential when given before or after the onset of Aß pathology.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Anticuerpos/uso terapéutico , Apolipoproteínas E/inmunología , Encéfalo/metabolismo , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/efectos de los fármacos , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/tratamiento farmacológico , Amiloidosis/metabolismo , Amiloidosis/patología , Animales , Encéfalo/efectos de los fármacos , Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Cojera Animal/tratamiento farmacológico , Cojera Animal/etiología , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Mutación/genética , Presenilina-1/genética
11.
Ann Neurol ; 75(2): 309-16, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24375753

RESUMEN

OBJECTIVE: Children with neurofibromatosis-1 (NF1) are at risk for developing numerous nervous system abnormalities, including cognitive problems and brain tumors (optic pathway glioma). Currently, there are few prognostic factors that predict clinical manifestations or outcomes in patients, even in families with an identical NF1 gene mutation. In this study, we leveraged Nf1 genetically engineered mice (GEM) to define the potential role of sex as a clinically relevant modifier of NF1-associated neuronal dysfunction. METHODS: Deidentified clinical data were analyzed to determine the impact of sex on optic glioma-associated visual decline in children with NF1. In addition, Nf1 GEM were employed as experimental platforms to investigate sexually dimorphic differences in learning/memory, visual acuity, retinal ganglion cell (RGC) death, and Nf1 protein (neurofibromin)-regulated signaling pathway function (Ras activity, cyclic adenosine monophosphate [cAMP], and dopamine levels). RESULTS: Female patients with NF1-associated optic glioma were twice as likely to undergo brain magnetic resonance imaging for visual symptoms and 3× more likely to require treatment for visual decline than their male counterparts. As such, only female Nf1 GEM exhibited a decrement in optic glioma-associated visual acuity, shorter RGC axons, and attenuated cAMP levels. In contrast, only male Nf1 GEM showed spatial learning/memory deficits, increased Ras activity, and reduced dopamine levels. INTERPRETATION: Collectively, these observations establish sex as a major prognostic factor underlying neuronal dysfunction in NF1, and suggest that sex should be considered when interpreting future preclinical and clinical study results.


Asunto(s)
Discapacidades para el Aprendizaje/etiología , Neurofibromatosis 1/complicaciones , Trastornos de la Visión/etiología , Animales , Encéfalo/patología , Niño , Dopamina/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/genética , Hipocampo/metabolismo , Humanos , Discapacidades para el Aprendizaje/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Glioma del Nervio Óptico/genética , Factores Sexuales , Percepción Espacial/fisiología
12.
J Neurosci ; 33(7): 2732-53, 2013 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23407934

RESUMEN

The immense molecular diversity of neurons challenges our ability to understand the genetic and cellular etiology of neuropsychiatric disorders. Leveraging knowledge from neurobiology may help parse the genetic complexity: identifying genes important for a circuit that mediates a particular symptom of a disease may help identify polymorphisms that contribute to risk for the disease as a whole. The serotonergic system has long been suspected in disorders that have symptoms of repetitive behaviors and resistance to change, including autism. We generated a bacTRAP mouse line to permit translational profiling of serotonergic neurons. From this, we identified several thousand serotonergic-cell expressed transcripts, of which 174 were highly enriched, including all known markers of these cells. Analysis of common variants near the corresponding genes in the AGRE collection implicated the RNA binding protein CELF6 in autism risk. Screening for rare variants in CELF6 identified an inherited premature stop codon in one of the probands. Subsequent disruption of Celf6 in mice resulted in animals exhibiting resistance to change and decreased ultrasonic vocalization as well as abnormal levels of serotonin in the brain. This work provides a reproducible and accurate method to profile serotonergic neurons under a variety of conditions and suggests a novel paradigm for gaining information on the etiology of psychiatric disorders.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/psicología , Perfilación de la Expresión Génica/métodos , Modificación Traduccional de las Proteínas/genética , Modificación Traduccional de las Proteínas/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Neuronas Serotoninérgicas/fisiología , Serotonina/fisiología , Animales , Conducta Animal/fisiología , Proteínas CELF , Estudio de Asociación del Genoma Completo , Humanos , Inmunohistoquímica , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Mutación/genética , Mutación/fisiología , Neurotransmisores/metabolismo , Polimorfismo Genético , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Ribosomas/genética , Ribosomas/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Olfato/fisiología , Conducta Social , Vocalización Animal/fisiología
13.
J Neurosci ; 33(31): 12887-97, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23904623

RESUMEN

Tau, a microtubule-associated protein, is implicated in the pathogenesis of Alzheimer's Disease (AD) in regard to both neurofibrillary tangle formation and neuronal network hyperexcitability. The genetic ablation of tau substantially reduces hyperexcitability in AD mouse lines, induced seizure models, and genetic in vivo models of epilepsy. These data demonstrate that tau is an important regulator of network excitability. However, developmental compensation in the genetic tau knock-out line may account for the protective effect against seizures. To test the efficacy of a tau reducing therapy for disorders with a detrimental hyperexcitability profile in adult animals, we identified antisense oligonucleotides that selectively decrease endogenous tau expression throughout the entire mouse CNS--brain and spinal cord tissue, interstitial fluid, and CSF--while having no effect on baseline motor or cognitive behavior. In two chemically induced seizure models, mice with reduced tau protein had less severe seizures than control mice. Total tau protein levels and seizure severity were highly correlated, such that those mice with the most severe seizures also had the highest levels of tau. Our results demonstrate that endogenous tau is integral for regulating neuronal hyperexcitability in adult animals and suggest that an antisense oligonucleotide reduction of tau could benefit those with epilepsy and perhaps other disorders associated with tau-mediated neuronal hyperexcitability.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Oligonucleótidos Antisentido/uso terapéutico , Convulsiones/prevención & control , Proteínas tau/genética , Factores de Edad , Animales , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Trastornos Neurológicos de la Marcha/tratamiento farmacológico , Trastornos Neurológicos de la Marcha/etiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Infusiones Intraventriculares , Ácido Láctico/metabolismo , Locomoción/genética , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microdiálisis , Pentilenotetrazol/toxicidad , Picrotoxina/toxicidad , Convulsiones/inducido químicamente , Convulsiones/genética , Proteínas tau/química , Proteínas tau/metabolismo
14.
Ann Neurol ; 73(2): 309-15, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23225063

RESUMEN

Children with neurofibromatosis type 1 (NF1) are prone to learning and behavioral abnormalities, including problems with spatial learning and attention. The molecular etiology for these deficits is unclear, as previous studies have implicated defective dopamine, cyclic adenosine monophosphate (cAMP), and Ras homeostasis. Using behavioral, electrophysiological, and primary culture, we now demonstrate that reduced dopamine signaling is responsible for cAMP-dependent defects in neuron function and learning. Collectively, these results establish defective dopaminergic function as a contributing factor underlying impaired spatial learning and memory in children and adults with NF1, and support the use of treatments that restore normal dopamine homeostasis for select individuals.


Asunto(s)
Dopamina/deficiencia , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/fisiopatología , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/fisiopatología , Animales , Atención/fisiología , AMP Cíclico/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiopatología , Discapacidades para el Aprendizaje/etiología , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurofibromatosis 1/complicaciones , Neurofibromina 1/genética , Transducción de Señal/fisiología , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/fisiopatología
15.
Biomolecules ; 14(3)2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38540675

RESUMEN

Brain cholesterol metabolic products include neurosteroids and oxysterols, which play important roles in cellular physiology. In neurons, the cholesterol oxidation product, 24S-hydroxycholesterol (24S-HC), is a regulator of signaling and transcription. Here, we examined the behavioral effects of 24S-HC loss, using global and cell-selective genetic deletion of the synthetic enzyme CYP46A1. Mice that are globally deficient in CYP46A1 exhibited hypoactivity at young ages and unexpected increases in conditioned fear memory. Despite strong reductions in hippocampal 24S-HC in mice with selective loss of CYP46A1 in VGLUT1-positive cells, behavioral effects were not recapitulated in these conditional knockout mice. Global knockout produced strong, developmentally dependent transcriptional effects on select cholesterol metabolism genes. These included paradoxical changes in Liver X Receptor targets. Again, conditional knockout was insufficient to recapitulate most changes. Overall, our results highlight the complex effects of 24S-HC in an in vivo setting that are not fully predicted by known mechanisms. The results also demonstrate that the complete inhibition of enzymatic activity may be needed for a detectable, therapeutically relevant impact on gene expression and behavior.


Asunto(s)
Colesterol , Hidroxicolesteroles , Ratones , Animales , Colesterol 24-Hidroxilasa/metabolismo , Hidroxicolesteroles/metabolismo , Colesterol/metabolismo , Hipocampo/metabolismo
16.
Am J Physiol Endocrinol Metab ; 304(12): E1331-7, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23592483

RESUMEN

Brain damage due to severe hypoglycemia occurs in insulin-treated people with diabetes. This study tests the hypothesis that chronic insulin therapy that normalizes elevated blood glucose in diabetic rats would be neuroprotective against brain damage induced by an acute episode of severe hypoglycemia. Male Sprague-Dawley rats were split into three groups: 1) control, non-diabetic; 2) STZ-diabetic; and 3) insulin-treated STZ-diabetic. After 3 wk of chronic treatment, unrestrained awake rats underwent acute hyperinsulinemic severe hypoglycemic (10-15 mg/dl) clamps for 1 h. Rats were subsequently analyzed for brain damage and cognitive function. Severe hypoglycemia induced 15-fold more neuronal damage in STZ-diabetic rats compared with nondiabetic rats. Chronic insulin treatment of diabetic rats, which nearly normalized glucose levels, markedly reduced neuronal damage induced by severe hypoglycemia. Fortunately, no cognitive defects associated with the hypoglycemia-induced brain damage were observed in any group. In conclusion, antecedent blood glucose control represents a major modifiable therapeutic intervention that can afford diabetic subjects neuroprotection against severe hypoglycemia-induced brain damage.


Asunto(s)
Encefalopatías/prevención & control , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemia/inducido químicamente , Insulina/farmacología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Encefalopatías/metabolismo , Enfermedad Crónica , Diabetes Mellitus Experimental/metabolismo , Técnica de Clampeo de la Glucosa , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Hipoglucemia/metabolismo , Hipoglucemia/patología , Hipoglucemiantes/farmacología , Masculino , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad
17.
J Inherit Metab Dis ; 36(3): 499-512, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22983812

RESUMEN

Mucopolysaccharidosis I (MPS I) is a lysosomal storage disease due to α-L-iduronidase (IDUA) deficiency that results in the accumulation of glycosaminoglycans (GAG). Systemic gene therapy to MPS I mice can reduce lysosomal storage in the brain, but few data are available regarding the effect upon behavioral function. We investigated the effect of gene therapy with a long-terminal-repeat (LTR)-intact retroviral vector or a self-inactivating (SIN) vector on behavioral function in MPS I mice. The LTR vector was injected intravenously to 6-week-old MPS I mice, and the SIN vector was given to neonatal or 6-week-old mice. Adult-LTR, neonatal-SIN, and adult-SIN-treated mice achieved serum IDUA activity of 235 ± 20 (84-fold normal), 127 ± 10, and 71 ± 7 U/ml, respectively. All groups had reduction in histochemical evidence of lysosomal storage in the brain, with the adult-LTR group showing the best response, while adult-LTR mice had reductions in lysosomal storage in the cristae of the vestibular system. Behavioral evaluation was performed at 8 months. Untreated MPS I mice had a markedly reduced ability to hold onto an inverted screen or climb down a pole. LTR-vector-treated mice had marked improvements on both of these tests, whereas neonatal-SIN mice showed improvement in the pole test. We conclude that both vectors can reduce brain disease in MPS I mice, with the LTR vector achieving higher serum IDUA levels and better correction. Vestibular abnormalities may contribute to mobility problems in patients with MPS I, and gene therapy may reduce symptoms.


Asunto(s)
Terapia Genética/métodos , Iduronidasa/genética , Trastornos Mentales/prevención & control , Enfermedad de la Neurona Motora/prevención & control , Mucopolisacaridosis I/terapia , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Encéfalo/enzimología , Encéfalo/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Trastornos Mentales/etiología , Trastornos Mentales/genética , Ratones , Ratones Endogámicos C57BL , Enfermedad de la Neurona Motora/etiología , Neuronas Motoras/fisiología , Mucopolisacaridosis I/complicaciones , Retroviridae
18.
Transl Stroke Res ; 14(5): 704-722, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36308676

RESUMEN

Posthemorrhagic hydrocephalus occurs in up to 30% of infants with high-grade intraventricular hemorrhage and is associated with the worst neurocognitive outcomes in preterm infants. The mechanisms of posthemorrhagic hydrocephalus after intraventricular hemorrhage are unknown; however, CSF levels of iron metabolic pathway proteins including hemoglobin have been implicated in its pathogenesis. Here, we develop an animal model of intraventricular hemorrhage using intraventricular injection of hemoglobin at post-natal day 4 that results in acute and chronic hydrocephalus, pathologic choroid plexus iron accumulation, and subsequent choroid plexus injury at post-natal days 5, 7, and 15. This model also results in increased expression of aquaporin-1, Na+/K+/Cl- cotransporter 1, and Na+/K+/ATPase on the apical surface of the choroid plexus 24 h post-intraventricular hemorrhage. We use this model to evaluate a clinically relevant treatment strategy for the prevention of neurological sequelae after intraventricular hemorrhage using intraventricular administration of the iron chelator deferoxamine at the time of hemorrhage. Deferoxamine treatment prevented posthemorrhagic hydrocephalus for up to 11 days after intraventricular hemorrhage and prevented the development of sensorimotor gating deficits. In addition, deferoxamine treatment facilitated acute iron clearance through the choroid plexus and subsequently reduced choroid plexus iron levels at 24 h with reversal of hemoglobin-induced aquaporin-1 upregulation on the apical surface of the choroid plexus. Intraventricular administration of deferoxamine at the time of intraventricular hemorrhage may be a clinically relevant treatment strategy for preventing posthemorrhagic hydrocephalus and likely acts through promoting iron clearance through the choroid plexus to prevent hemoglobin-induced injury.


Asunto(s)
Acuaporinas , Hidrocefalia , Recién Nacido , Humanos , Animales , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Hierro , Deferoxamina/uso terapéutico , Recien Nacido Prematuro , Hidrocefalia/etiología , Hidrocefalia/prevención & control , Hidrocefalia/patología , Hemorragia Cerebral/metabolismo , Hemoglobinas/metabolismo , Acuaporinas/metabolismo
19.
J Neurosci ; 31(27): 9945-57, 2011 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-21734286

RESUMEN

Globoid-cell leukodystrophy (GLD) is an inherited demyelinating disease caused by the deficiency of the lysosomal enzyme galactosylceramidase (GALC). A previous study in the murine model of GLD (twitcher) demonstrated a dramatic synergy between CNS-directed adeno-associated virus 2/5 (AAV2/5) gene therapy and myeloreductive bone marrow transplantation (BMT). However, the mechanism by which these two disparate therapeutic approaches synergize is not clear. In addition, the therapeutic efficacy may have been limited since the CNS-directed gene therapy was restricted to the forebrain and thalamus. In the current study, intrathecal and intracerebellar injections were added to the therapeutic regimen and the mechanism of synergy between BMT and gene therapy was determined. Although AAV2/5 alone provided supraphysiological levels of GALC activity and reduced psychosine levels in both the brain and spinal cord, it significantly increased CNS inflammation. Bone marrow transplantation alone provided essentially no GALC activity to the CNS and did not reduce psychosine levels. When AAV2/5 is combined with BMT, there are sustained improvements in motor function and the median life span is increased to 123 d (range, 92-282 d) compared with 41 d in the untreated twitcher mice. Interestingly, addition of BMT virtually eliminates both the disease and AAV2/5-associated inflammatory response. These data suggest that the efficacy of AAV2/5-mediated gene therapy is limited by the associated inflammatory response and BMT synergizes with AAV2/5 by modulating inflammation.


Asunto(s)
Trasplante de Médula Ósea/métodos , Encéfalo/metabolismo , Terapia Genética/métodos , Inflamación/terapia , Leucodistrofia de Células Globoides/terapia , Médula Espinal/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Dependovirus/genética , Imagen de Difusión Tensora/métodos , Modelos Animales de Enfermedad , Citometría de Flujo/métodos , Galactosilceramidasa/biosíntesis , Galactosilceramidasa/deficiencia , Vectores Genéticos/fisiología , Indoles , Inflamación/etiología , Estimación de Kaplan-Meier , Leucodistrofia de Células Globoides/complicaciones , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/patología , Longevidad/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido Peryódico , Psicosina/metabolismo , Temblor/etiología
20.
Hum Mol Genet ; 19(22): 4515-28, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20826448

RESUMEN

Learning and behavioral abnormalities are among the most common clinical problems in children with the neurofibromatosis-1 (NF1) inherited cancer syndrome. Recent studies using Nf1 genetically engineered mice (GEM) have been instructive for partly elucidating the cellular and molecular defects underlying these cognitive deficits; however, no current model has shed light on the more frequently encountered attention system abnormalities seen in children with NF1. Using an Nf1 optic glioma (OPG) GEM model, we report novel defects in non-selective and selective attention without an accompanying hyperactivity phenotype. Specifically, Nf1 OPG mice exhibit reduced rearing in response to novel objects and environmental stimuli. Similar to children with NF1, the attention system dysfunction in these mice is reversed by treatment with methylphenidate (MPH), suggesting a defect in brain catecholamine homeostasis. We further demonstrate that this attention system abnormality is the consequence of reduced dopamine (DA) levels in the striatum, which is normalized following either MPH or l-dopa administration. The reduction in striatal DA levels in Nf1 OPG mice is associated with reduced striatal expression of tyrosine hydroxylase, the rate-limited enzyme in DA synthesis, without any associated dopaminergic cell loss in the substantia nigra. Moreover, we demonstrate a cell-autonomous defect in Nf1+/- dopaminergic neuron growth cone areas and neurite extension in vitro, which results in decreased dopaminergic cell projections to the striatum in Nf1 OPG mice in vivo. Collectively, these data establish abnormal DA homeostasis as the primary biochemical defect underlying the attention system dysfunction in Nf1 GEM relevant to children with NF1.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Atención , Dopamina/metabolismo , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Animales , Encéfalo/metabolismo , Niño , Cuerpo Estriado/metabolismo , Dopamina/genética , Genes de Neurofibromatosis 1 , Humanos , Levodopa/genética , Levodopa/metabolismo , Metilfenidato/metabolismo , Metilfenidato/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Actividad Motora/genética , Neurofibromatosis 1/enzimología , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Neuronas/metabolismo , Glioma del Nervio Óptico/genética , Glioma del Nervio Óptico/metabolismo , Sustancia Negra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA