Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 145(6): 863-74, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21640374

RESUMEN

Metabolites in the kynurenine pathway, generated by tryptophan degradation, are thought to play an important role in neurodegenerative disorders, including Alzheimer's and Huntington's diseases. In these disorders, glutamate receptor-mediated excitotoxicity and free radical formation have been correlated with decreased levels of the neuroprotective metabolite kynurenic acid. Here, we describe the synthesis and characterization of JM6, a small-molecule prodrug inhibitor of kynurenine 3-monooxygenase (KMO). Chronic oral administration of JM6 inhibits KMO in the blood, increasing kynurenic acid levels and reducing extracellular glutamate in the brain. In a transgenic mouse model of Alzheimer's disease, JM6 prevents spatial memory deficits, anxiety-related behavior, and synaptic loss. JM6 also extends life span, prevents synaptic loss, and decreases microglial activation in a mouse model of Huntington's disease. These findings support a critical link between tryptophan metabolism in the blood and neurodegeneration, and they provide a foundation for treatment of neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Huntington/tratamiento farmacológico , Ácido Quinurénico/análisis , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Tiazoles/uso terapéutico , Administración Oral , Enfermedad de Alzheimer/fisiopatología , Animales , Química Encefálica , Modelos Animales de Enfermedad , Femenino , Humanos , Ácido Quinurénico/sangre , Masculino , Ratones , Ratones Transgénicos , Sulfonamidas/administración & dosificación , Tiazoles/administración & dosificación
2.
J Neurosci ; 36(7): 2316-22, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26888940

RESUMEN

Apolipoprotein E4 (apoE4) is the major genetic risk factor for Alzheimer's disease (AD). However, the underlying mechanisms are still poorly understood. We previously reported that female apoE4 knock-in (KI) mice had an age-dependent decline in hilar GABAergic interneurons that correlated with the extent of learning and memory deficits, as determined by Morris water maze (MWM), in aged mice. Enhancing GABA signaling by treating aged apoE4-KI mice with the GABAA receptor potentiator pentobarbital (PB) for 4 weeks before and during MWM rescued the learning and memory deficits. Here, we report that withdrawal of PB treatment for 2 weeks before MWM abolished the rescue in aged apoE4-KI mice, suggesting the importance of continuously enhancing GABA signaling in the rescue. However, treating apoE4-KI mice during middle adulthood (9-11 months of age) with PB for 6 weeks prevented age-dependent hilar GABAergic interneuron decline and learning and memory deficits, when examined at 16 month of age. These data imply that increasing inhibitory tone after substantial GABAergic interneuron loss may be an effective symptomatic, but not a disease-modifying, treatment for AD related to apoE4, whereas a similar intervention before substantial interneuron loss could be a disease-modifying therapeutic. SIGNIFICANCE STATEMENT: We previously reported that female apoE4-KI mice had an age-dependent decline in hilar GABAergic interneurons that correlated with the extent of cognitive deficits in aged mice. The current study demonstrates that enhancing GABA signaling by treating aged apoE4-KI mice with a GABAA receptor potentiator pentobarbital (PB) before and during behavioral tests rescued the cognitive deficits; but withdrawal of PB treatment for 2 weeks before the tests abolished the rescue, suggesting the importance of continuously enhancing GABA signaling. However, treating apoE4-KI mice during middle adulthood with PB for a short period of time prevented age-dependent hilar GABAergic interneuron decline and cognitive deficits late in life, suggesting early intervention by enhancing GABA signaling as a potential strategy to prevent AD related to apoE4.


Asunto(s)
Apolipoproteína E4/genética , Agonistas del GABA/farmacología , Interneuronas/efectos de los fármacos , Discapacidades para el Aprendizaje/prevención & control , Trastornos de la Memoria/prevención & control , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/genética , Envejecimiento/genética , Envejecimiento/psicología , Animales , Femenino , Técnicas de Sustitución del Gen , Discapacidades para el Aprendizaje/genética , Aprendizaje por Laberinto , Trastornos de la Memoria/genética , Ratones , Ratones Endogámicos C57BL , Pentobarbital/farmacología , Receptores de GABA-A/efectos de los fármacos , Transducción de Señal/genética
3.
Proc Natl Acad Sci U S A ; 108(10): 4236-41, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21368138

RESUMEN

Apolipoprotein (apo) E4 is the major known genetic risk factor for Alzheimer's disease (AD). We have shown in vitro and in vivo that apoE4 preferentially undergoes aberrant cleavage in neurons, yielding neurotoxic C-terminal-truncated fragments. To study the effect of these fragments on amyloid-ß (Aß) clearance/deposition and their potential synergy with Aß in eliciting neuronal and behavioral deficits, we cross-bred transgenic mice expressing apoE3, apoE4, or apoE4(Δ272-299) with mice expressing human amyloid protein precursor (APP) harboring familial AD mutations (hAPP(FAD)). At 6-8 mo of age, hAPP(FAD) mice expressing apoE3 or apoE4 had lower levels of hippocampal Aß (94% and 89%, respectively) and less Aß deposition (89% and 87%) than hAPP(FAD) mice without apoE, whereas hAPP(FAD) mice expressing mouse apoE had higher Aß levels. Thus, human apoE stimulates Aß clearance, but mouse apoE does not. Expression of apoE4(Δ272-299) reduced total Aß levels by only 63% and Aß deposition by 46% compared with hAPP(FAD) mice without apoE. Unlike apoE3 and apoE4, the C-terminal-truncated apoE4 bound poorly with Aß peptides, leading to decreased Aß clearance and increased Aß deposition. Despite their lower levels of Aß and Aß deposition, hAPP(FAD)/apoE4(Δ272-299) mice accumulated pathogenic Aß oligomers and displayed neuronal and behavioral deficits similar to or more severe than those in hAPP(FAD) mice. Thus, the C-terminal-truncated apoE4 fragment inefficiently clears Aß peptides and acts in concert with low levels of Aß to elicit neuronal and behavioral deficits in mice.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/fisiología , Conducta Animal , Neuronas/fisiología , Animales , Apolipoproteína E4/metabolismo , Humanos , Ratones , Unión Proteica
5.
Invest Ophthalmol Vis Sci ; 64(2): 3, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36729444

RESUMEN

Purpose: C1q and the classical complement cascade are key regulators of synaptic pruning, and their aberrant activation has been implicated in neurodegenerative ophthalmic diseases including geographic atrophy and glaucoma. The antigen-binding fragment antibody ANX007 specifically recognizes globular head groups of C1q to block substrate binding and functionally inhibit classical complement cascade activation. ANX007 was assessed in nonclinical studies of biodistribution and C1q target engagement in the eye following intravitreal (IVT) administration in cynomolgus monkeys. Methods: Female juvenile cynomolgus monkeys (n = 12) received a single bilateral dose of 1 or 5 mg ANX007/eye, with vitreous and non-perfused tissue samples collected approximately 4 weeks later. In a separate study, male (n = 6/5) and female (n = 6/5) animals received repeat bilateral dosing of 1, 2.5, or 5 mg ANX007/eye on days 1 and 29, with aqueous and vitreous collections on day 44 or day 59. Tissues from the 5 mg/eye repeat-dose group were perfused, and retina, choroid, and optic nerve samples were collected approximately 2 and 4 weeks post-last dose. Results: Following a single dose of ANX007, vitreous levels of free drug were measurable through 4 weeks at both the 1 and 5 mg dose levels, with approximately 3-day half-life. With repeat dose of 5 mg/eye, free-ANX007 was measurable 4 weeks post-last dose in perfused retina and choroid and up to approximately 2 weeks post-last dose in optic nerve. There was a strong correlation between C1q target engagement and free drug levels in aqueous and vitreous humors and retinal tissue. Conclusions: Following IVT administration, ANX007 distributes to sites within the retina that are relevant to neurodegenerative ophthalmic disease with clear evidence of C1q target engagement. Based on its mechanism of action inhibiting C1q and its downstream activity, ANX007 is predicted to mitigate tissue damage driven by classical complement activation in the retina. These data support further clinical evaluation of ANX007.


Asunto(s)
Retina , Cuerpo Vítreo , Animales , Masculino , Femenino , Macaca fascicularis , Distribución Tisular , Retina/metabolismo , Cuerpo Vítreo/metabolismo , Fragmentos Fab de Inmunoglobulinas
6.
J Neurosci ; 30(41): 13707-17, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20943911

RESUMEN

Apolipoprotein E4 (apoE4) is the major genetic risk factor for Alzheimer's disease. However, the underlying mechanisms are unclear. We found that female apoE4 knock-in (KI) mice had an age-dependent decrease in hilar GABAergic interneurons that correlated with the extent of learning and memory deficits, as determined in the Morris water maze, in aged mice. Treating apoE4-KI mice with daily peritoneal injections of the GABA(A) receptor potentiator pentobarbital at 20 mg/kg for 4 weeks rescued the learning and memory deficits. In neurotoxic apoE4 fragment transgenic mice, hilar GABAergic interneuron loss was even more pronounced and also correlated with the extent of learning and memory deficits. Neurodegeneration and tauopathy occurred earliest in hilar interneurons in apoE4 fragment transgenic mice; eliminating endogenous Tau prevented hilar GABAergic interneuron loss and the learning and memory deficits. The GABA(A) receptor antagonist picrotoxin abolished this rescue, while pentobarbital rescued learning deficits in the presence of endogenous Tau. Thus, apoE4 causes age- and Tau-dependent impairment of hilar GABAergic interneurons, leading to learning and memory deficits in mice. Consequently, reducing Tau and enhancing GABA signaling are potential strategies to treat or prevent apoE4-related Alzheimer's disease.


Asunto(s)
Apolipoproteína E4/metabolismo , Giro Dentado/metabolismo , Interneuronas/fisiología , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Proteínas tau/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Apolipoproteína E4/genética , Células Cultivadas , Giro Dentado/efectos de los fármacos , Giro Dentado/fisiopatología , Electrofisiología , Femenino , Inmunohistoquímica , Interneuronas/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Pentobarbital/administración & dosificación , Terminales Presinápticos/fisiología , Estadísticas no Paramétricas
7.
Science ; 373(6560): eabj2685, 2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34516796

RESUMEN

Although traumatic brain injury (TBI) acutely disrupts the cortex, most TBI-related disabilities reflect secondary injuries that accrue over time. The thalamus is a likely site of secondary damage because of its reciprocal connections with the cortex. Using a mouse model of mild TBI (mTBI), we found a chronic increase in C1q expression specifically in the corticothalamic system. Increased C1q expression colocalized with neuron loss and chronic inflammation and correlated with disruption in sleep spindles and emergence of epileptic activities. Blocking C1q counteracted these outcomes, suggesting that C1q is a disease modifier in mTBI. Single-nucleus RNA sequencing demonstrated that microglia are a source of thalamic C1q. The corticothalamic circuit could thus be a new target for treating TBI-related disabilities.


Asunto(s)
Lesiones Encefálicas/complicaciones , Complemento C1q/fisiología , Fases del Sueño , Trastornos del Sueño-Vigilia/etiología , Trastornos del Sueño-Vigilia/fisiopatología , Tálamo/fisiopatología , Animales , Lesiones Encefálicas/fisiopatología , Complemento C1q/genética , Modelos Animales de Enfermedad , Epilepsia/fisiopatología , Ratones , Microglía/metabolismo , Tálamo/metabolismo
8.
PLoS One ; 7(12): e53569, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300939

RESUMEN

Apolipoprotein (apo) E4 is the major genetic risk factor for Alzheimer's disease (AD). ApoE4 has sex-dependent effects, whereby the risk of developing AD is higher in apoE4-expressing females than males. However, the mechanism underlying the sex difference, in relation to apoE4, is unknown. Previous findings indicate that apoE4 causes age-dependent impairments of hilar GABAergic interneurons in female mice, leading to learning and memory deficits. Here, we investigate whether the detrimental effects of apoE4 on hilar GABAergic interneurons are sex-dependent using apoE knock-in (KI) mice across different ages. We found that in female apoE-KI mice, there was an age-dependent depletion of hilar GABAergic interneurons, whereby GAD67- or somatostatin-positive--but not NPY- or parvalbumin-positive-interneuron loss was exacerbated by apoE4. Loss of these neuronal populations was correlated with the severity of spatial learning deficits at 16 months of age in female apoE4-KI mice; however, this effect was not observed in female apoE3-KI mice. In contrast, we found an increase in the numbers of hilar GABAergic interneurons with advancing age in male apoE-KI mice, regardless of apoE genotype. Moreover, male apoE-KI mice showed a consistent ratio of hilar inhibitory GABAergic interneurons to excitatory mossy cells approximating 1.5 that is independent of apoE genotype and age, whereas female apoE-KI mice exhibited an age-dependent decrease in this ratio, which was exacerbated by apoE4. Interestingly, there are no apoE genotype effects on GABAergic interneurons in the CA1 and CA3 subregions of the hippocampus as well as the entorhinal and auditory cortexes. These findings suggest that the sex-dependent effects of apoE4 on developing AD is in part attributable to inherent sex-based differences in the numbers of hilar GABAergic interneurons, which is further modulated by apoE genotype.


Asunto(s)
Apolipoproteína E4/genética , Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Interneuronas/metabolismo , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/genética , Factores de Edad , Animales , Apolipoproteína E4/metabolismo , Técnicas de Sustitución del Gen , Trastornos de la Memoria/metabolismo , Ratones , Ratones Transgénicos , Factores Sexuales
9.
Cell Stem Cell ; 11(1): 100-9, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22683203

RESUMEN

The generation of induced pluripotent stem cells (iPSCs) and induced neuronal cells (iNCs) from somatic cells provides new avenues for basic research and potential transplantation therapies for neurological diseases. However, clinical applications must consider the risk of tumor formation by iPSCs and the inability of iNCs to self-renew in culture. Here we report the generation of induced neural stem cells (iNSCs) from mouse and human fibroblasts by direct reprogramming with a single factor, Sox2. iNSCs express NSC markers and resemble wild-type NSCs in their morphology, self-renewal, ability to form neurospheres, and gene expression profiles. Cloned iNSCs differentiate into several types of mature neurons, as well as astrocytes and oligodendrocytes, indicating multipotency. Implanted iNSCs can survive and integrate in mouse brains and, unlike iPSC-derived NSCs, do not generate tumors. Thus, self-renewable and multipotent iNSCs without tumorigenic potential can be generated directly from fibroblasts by reprogramming.


Asunto(s)
Reprogramación Celular/genética , Fibroblastos/citología , Células Madre Multipotentes/citología , Células-Madre Neurales/citología , Factores de Transcripción SOXB1/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Embrión de Mamíferos/citología , Feto/citología , Fibroblastos/metabolismo , Humanos , Ratones , Células Madre Multipotentes/metabolismo , Neoplasias/patología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo
10.
PLoS One ; 7(7): e40555, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792368

RESUMEN

BACKGROUND: Although extensive research has demonstrated the importance of excitatory granule neurons in the dentate gyrus of the hippocampus in normal learning and memory and in the pathogenesis of amnesia in Alzheimer's disease (AD), the role of hilar GABAergic inhibitory interneurons, which control the granule neuron activity, remains unclear. METHODOLOGY AND PRINCIPAL FINDINGS: We explored the function of hilar GABAergic interneurons in spatial learning and memory by inhibiting their activity through Cre-dependent viral expression of enhanced halorhodopsin (eNpHR3.0)--a light-driven chloride pump. Hilar GABAergic interneuron-specific expression of eNpHR3.0 was achieved by bilaterally injecting adeno-associated virus containing a double-floxed inverted open-reading frame encoding eNpHR3.0 into the hilus of the dentate gyrus of mice expressing Cre recombinase under the control of an enhancer specific for GABAergic interneurons. In vitro and in vivo illumination with a yellow laser elicited inhibition of hilar GABAergic interneurons and consequent activation of dentate granule neurons, without affecting pyramidal neurons in the CA3 and CA1 regions of the hippocampus. We found that optogenetic inhibition of hilar GABAergic interneuron activity impaired spatial learning and memory retrieval, without affecting memory retention, as determined in the Morris water maze test. Importantly, optogenetic inhibition of hilar GABAergic interneuron activity did not alter short-term working memory, motor coordination, or exploratory activity. CONCLUSIONS AND SIGNIFICANCE: Our findings establish a critical role for hilar GABAergic interneuron activity in controlling spatial learning and memory retrieval and provide evidence for the potential contribution of GABAergic interneuron impairment to the pathogenesis of amnesia in AD.


Asunto(s)
Giro Dentado/metabolismo , Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Aprendizaje/fisiología , Memoria/fisiología , Animales , Giro Dentado/citología , Dependovirus/genética , Genes Reporteros , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Memoria a Corto Plazo/fisiología , Ratones , Ratones Transgénicos , Desempeño Psicomotor
11.
Cell Stem Cell ; 5(6): 634-45, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19951691

RESUMEN

Apolipoprotein (apo) E, a polymorphic protein with three isoforms (apoE2, apoE3, and apoE4), is essential for lipid homeostasis. Carriers of apoE4 are at higher risk for developing Alzheimer's disease. We have investigated adult neurogenesis in mice with knockout (KO) for apoE or with knockin (KI) alleles for human apoE3 or apoE4, and we report that neurogenesis is reduced in both apoE-KO and apoE4-KI mice. In apoE-KO mice, increased BMP signaling promoted glial differentiation at the expense of neurogenesis. In contrast, in apoE4-KI mice, presynaptic GABAergic input-mediated maturation of newborn neurons was diminished. Tau phosphorylation, an Alzheimer's disease characteristic, and levels of neurotoxic apoE fragments were both elevated in apoE4-KI hippocampal neurons concomitant with decreased GABAergic interneuron survival. Potentiating GABAergic signaling restored neuronal maturation and neurogenesis in apoE4-KI mice to normal levels. These findings suggest that GABAergic signaling can be targeted to mitigate the deleterious effects of apoE4 on neurogenesis.


Asunto(s)
Células Madre Adultas/metabolismo , Enfermedad de Alzheimer/metabolismo , Apolipoproteínas/metabolismo , Neuroglía/metabolismo , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Animales Recién Nacidos , Apolipoproteínas/genética , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Agonistas del GABA/administración & dosificación , Técnicas de Sustitución del Gen , Hipocampo/patología , Humanos , Interneuronas/metabolismo , Interneuronas/patología , Ratones , Ratones Noqueados , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuroglía/efectos de los fármacos , Neuroglía/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas tau/metabolismo
12.
J Biol Chem ; 281(28): 19720-31, 2006 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-16704978

RESUMEN

Transmitter release at synapses between nerve cells is spatially restricted to active zones, where synaptic vesicle docking, priming, and Ca2+-dependent fusion take place in a temporally highly coordinated manner. Munc13s are essential for priming synaptic vesicles to a fusion competent state, and their specific active zone localization contributes to the active zone restriction of transmitter release and the speed of excitation-secretion coupling. However, the molecular mechanism of the active zone recruitment of Munc13s is not known. We show here that the active zone recruitment of Munc13 isoforms Munc13-1 and ubMunc13-2 is regulated by their binding to the Rab3A-interacting molecule RIM1alpha, a key determinant of long term potentiation of synaptic transmission at mossy fiber synapses in the hippocampus. We identify a single point mutation in Munc13-1 and ubMunc13-2 (I121N) that, depending on the type of assay used, strongly perturbs or abolishes RIM1alpha binding in vitro and in cultured fibroblasts, and we demonstrate that RIM1alpha binding-deficient ubMunc13-2(I121) is not efficiently recruited to synapses. Moreover, the levels of Munc13-1 and ubMunc13-2 levels are decreased in RIM1alpha-deficient brain, and Munc13-1 is not properly enriched at active zones of mossy fiber terminals of the mouse hippocampus if RIM1alpha is absent. We conclude that one function of the Munc13/RIM1alpha interaction is the active zone recruitment of Munc13-1 and ubMunc13-2.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína de Unión al GTP rab3A/metabolismo , Secuencia de Aminoácidos , Animales , Hipocampo/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Neuronas/metabolismo , Ratas , Homología de Secuencia de Aminoácido , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA