Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell ; 149(3): 708-21, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22541439

RESUMEN

Alzheimer's disease (AD) results in cognitive decline and altered network activity, but the mechanisms are unknown. We studied human amyloid precursor protein (hAPP) transgenic mice, which simulate key aspects of AD. Electroencephalographic recordings in hAPP mice revealed spontaneous epileptiform discharges, indicating network hypersynchrony, primarily during reduced gamma oscillatory activity. Because this oscillatory rhythm is generated by inhibitory parvalbumin (PV) cells, network dysfunction in hAPP mice might arise from impaired PV cells. Supporting this hypothesis, hAPP mice and AD patients had decreased levels of the interneuron-specific and PV cell-predominant voltage-gated sodium channel subunit Nav1.1. Restoring Nav1.1 levels in hAPP mice by Nav1.1-BAC expression increased inhibitory synaptic activity and gamma oscillations and reduced hypersynchrony, memory deficits, and premature mortality. We conclude that reduced Nav1.1 levels and PV cell dysfunction critically contribute to abnormalities in oscillatory rhythms, network synchrony, and memory in hAPP mice and possibly in AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Interneuronas/metabolismo , Aprendizaje , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.1 , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Canales de Sodio/metabolismo , Sinapsis
2.
J Neurosci ; 38(15): 3680-3688, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29540553

RESUMEN

Hyperacetylation of tau has been implicated in neurodegeneration and cognitive decline in tauopathy brains. The nicotinamide adenosine dinucleotide-dependent class-III protein deacetylase SIRT1 is one of the major enzymes involved in removal of acetyl groups from tau in vitro However, whether SIRT1 regulates acetylation of pathogenic tau and ameliorates tau-mediated pathogenesis remains unclear. Here, we report deacetylating activity of SIRT1 for acetylated Lys174 (K174) of tau in tauP301S transgenic mice with a brain-specific SIRT1 deletion. We show that SIRT1 deficiency leads to exacerbation of premature mortality, synapse loss, and behavioral disinhibition in tauP301S transgenic mice of both sexes. By contrast, SIRT1 overexpression by stereotaxic delivery of adeno-associated virus that encodes SIRT1 into the hippocampus reduces acetylated K174 tau. Furthermore, SIRT1 overexpression significantly attenuates the spread of tau pathology into anatomically connected brain regions of tauP301S transgenic mice of both sexes. These findings suggest the functional importance of SIRT1 in regulating pathogenic tau acetylation and in suppressing the spread of tau pathology in vivoSIGNIFICANCE STATEMENT In neurodegenerative disorders with inclusions of microtubule-associated protein tau, aberrant lysine acetylation of tau plays critical roles in promoting tau accumulation and toxicity. Identifying strategies to deacetylate tau could interfere with disease progression; however, little is known about how pathogenic tau is deacetylated in vivo Here we show that the protein deacetylase SIRT1 reduces tau acetylation in a mouse model of neurodegeneration. SIRT1 deficiency in the brain aggravates synapse loss and behavioral disinhibition, and SIRT1 overexpression ameliorates propagation of tau pathology.


Asunto(s)
Sirtuina 1/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Acetilación , Animales , Femenino , Células HEK293 , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Sirtuina 1/genética , Transmisión Sináptica , Tauopatías/patología , Tauopatías/fisiopatología
3.
Nature ; 469(7328): 47-52, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21113149

RESUMEN

Amyloid-ß oligomers may cause cognitive deficits in Alzheimer's disease by impairing neuronal NMDA-type glutamate receptors, whose function is regulated by the receptor tyrosine kinase EphB2. Here we show that amyloid-ß oligomers bind to the fibronectin repeats domain of EphB2 and trigger EphB2 degradation in the proteasome. To determine the pathogenic importance of EphB2 depletions in Alzheimer's disease and related models, we used lentiviral constructs to reduce or increase neuronal expression of EphB2 in memory centres of the mouse brain. In nontransgenic mice, knockdown of EphB2 mediated by short hairpin RNA reduced NMDA receptor currents and impaired long-term potentiation in the dentate gyrus, which are important for memory formation. Increasing EphB2 expression in the dentate gyrus of human amyloid precursor protein transgenic mice reversed deficits in NMDA receptor-dependent long-term potentiation and memory impairments. Thus, depletion of EphB2 is critical in amyloid-ß-induced neuronal dysfunction. Increasing EphB2 levels or function could be beneficial in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Cognición/fisiología , Receptor EphB2/deficiencia , Receptor EphB2/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular , Células Cultivadas , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Humanos , Potenciación a Largo Plazo , Memoria/fisiología , Ratones , Ratones Transgénicos , Plasticidad Neuronal , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Receptor EphB2/química , Receptor EphB2/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo
4.
J Neurosci ; 35(2): 807-18, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25589773

RESUMEN

Aging is the predominant risk factor for neurodegenerative diseases. One key phenotype as the brain ages is an aberrant innate immune response characterized by proinflammation. However, the molecular mechanisms underlying aging-associated proinflammation are poorly defined. Whether chronic inflammation plays a causal role in cognitive decline in aging and neurodegeneration has not been established. Here we report a mechanistic link between chronic inflammation and aging microglia and a causal role of aging microglia in neurodegenerative cognitive deficits. We showed that SIRT1 is reduced with the aging of microglia and that microglial SIRT1 deficiency has a causative role in aging- or tau-mediated memory deficits via IL-1ß upregulation in mice. Interestingly, the selective activation of IL-1ß transcription by SIRT1 deficiency is likely mediated through hypomethylating the specific CpG sites on IL-1ß proximal promoter. In humans, hypomethylation of IL-1ß is strongly associated with chronological age and with elevated IL-1ß transcription. Our findings reveal a novel epigenetic mechanism in aging microglia that contributes to cognitive deficits in aging and neurodegenerative diseases.


Asunto(s)
Envejecimiento/metabolismo , Cognición , Epigénesis Genética , Interleucina-1beta/metabolismo , Microglía/metabolismo , Sirtuina 1/metabolismo , Animales , Estudios de Casos y Controles , Metilación de ADN , Humanos , Interleucina-1beta/genética , Ratones , Sirtuina 1/deficiencia , Sirtuina 1/genética , Tauopatías/metabolismo , Regulación hacia Arriba
5.
Proc Natl Acad Sci U S A ; 109(42): E2895-903, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-22869752

RESUMEN

In light of the rising prevalence of Alzheimer's disease (AD), new strategies to prevent, halt, and reverse this condition are needed urgently. Perturbations of brain network activity are observed in AD patients and in conditions that increase the risk of developing AD, suggesting that aberrant network activity might contribute to AD-related cognitive decline. Human amyloid precursor protein (hAPP) transgenic mice simulate key aspects of AD, including pathologically elevated levels of amyloid-ß peptides in brain, aberrant neural network activity, remodeling of hippocampal circuits, synaptic deficits, and behavioral abnormalities. Whether these alterations are linked in a causal chain remains unknown. To explore whether hAPP/amyloid-ß-induced aberrant network activity contributes to synaptic and cognitive deficits, we treated hAPP mice with different antiepileptic drugs. Among the drugs tested, only levetiracetam (LEV) effectively reduced abnormal spike activity detected by electroencephalography. Chronic treatment with LEV also reversed hippocampal remodeling, behavioral abnormalities, synaptic dysfunction, and deficits in learning and memory in hAPP mice. Our findings support the hypothesis that aberrant network activity contributes causally to synaptic and cognitive deficits in hAPP mice. LEV might also help ameliorate related abnormalities in people who have or are at risk for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Anticonvulsivantes/farmacología , Trastornos del Conocimiento/tratamiento farmacológico , Cognición/efectos de los fármacos , Red Nerviosa/efectos de los fármacos , Piracetam/análogos & derivados , Sinapsis/efectos de los fármacos , Enfermedad de Alzheimer/complicaciones , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Anticonvulsivantes/sangre , Anticonvulsivantes/uso terapéutico , Western Blotting , Trastornos del Conocimiento/etiología , Electroencefalografía , Humanos , Inmunohistoquímica , Levetiracetam , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Red Nerviosa/fisiopatología , Piracetam/sangre , Piracetam/farmacología , Piracetam/uso terapéutico
6.
J Neurosci ; 31(2): 700-11, 2011 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-21228179

RESUMEN

Alzheimer's disease (AD), the most common neurodegenerative disorder, is a growing public health problem and still lacks effective treatments. Recent evidence suggests that microtubule-associated protein tau may mediate amyloid-ß peptide (Aß) toxicity by modulating the tyrosine kinase Fyn. We showed previously that tau reduction prevents, and Fyn overexpression exacerbates, cognitive deficits in human amyloid precursor protein (hAPP) transgenic mice overexpressing Aß. However, the mechanisms by which Aß, tau, and Fyn cooperate in AD-related pathogenesis remain to be fully elucidated. Here we examined the synaptic and network effects of this pathogenic triad. Tau reduction prevented cognitive decline induced by synergistic effects of Aß and Fyn. Tau reduction also prevented synaptic transmission and plasticity deficits in hAPP mice. Using electroencephalography to examine network effects, we found that tau reduction prevented spontaneous epileptiform activity in multiple lines of hAPP mice. Tau reduction also reduced the severity of spontaneous and chemically induced seizures in mice overexpressing both Aß and Fyn. To better understand these protective effects, we recorded whole-cell currents in acute hippocampal slices from hAPP mice with and without tau. hAPP mice with tau had increased spontaneous and evoked excitatory currents, reduced inhibitory currents, and NMDA receptor dysfunction. Tau reduction increased inhibitory currents and normalized excitation/inhibition balance and NMDA receptor-mediated currents in hAPP mice. Our results indicate that Aß, tau, and Fyn jointly impair synaptic and network function and suggest that disrupting the copathogenic relationship between these factors could be of therapeutic benefit.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/fisiología , Trastornos del Conocimiento/fisiopatología , Red Nerviosa/fisiología , Proteínas Proto-Oncogénicas c-fyn/fisiología , Sinapsis/fisiología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/mortalidad , Animales , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/psicología , Modelos Animales de Enfermedad , Electroencefalografía , Femenino , Hipocampo/fisiopatología , Técnicas In Vitro , Masculino , Ratones , Ratones Mutantes , Plasticidad Neuronal , Convulsiones/metabolismo , Convulsiones/fisiopatología , Especificidad de la Especie , Transmisión Sináptica , Proteínas tau/genética
7.
J Neurosci ; 30(1): 372-81, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20053918

RESUMEN

Previous studies suggested that cleavage of the amyloid precursor protein (APP) at aspartate residue 664 by caspases may play a key role in the pathogenesis of Alzheimer's disease. Mutation of this site (D664A) prevents caspase cleavage and the generation of the C-terminal APP fragments C31 and Jcasp, which have been proposed to mediate amyloid-beta (Abeta) neurotoxicity. Here we compared human APP transgenic mice with (B254) and without (J20) the D664A mutation in a battery of tests. Before Abeta deposition, hAPP-B254 and hAPP-J20 mice had comparable hippocampal levels of Abeta(1-42). At 2-3 or 5-7 months of age, hAPP-B254 and hAPP-J20 mice had similar abnormalities relative to nontransgenic mice in spatial and nonspatial learning and memory, elevated plus maze performance, electrophysiological measures of synaptic transmission and plasticity, and levels of synaptic activity-related proteins. Thus, caspase cleavage of APP at position D664 and generation of C31 do not play a critical role in the development of these abnormalities.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Caspasas/metabolismo , Modelos Animales de Enfermedad , Neuronas/metabolismo , Desempeño Psicomotor/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Humanos , Hidrólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neuronas/patología
8.
Proc Natl Acad Sci U S A ; 105(20): 7333-8, 2008 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-18480251

RESUMEN

Estrogens act within the ventromedial nucleus of the hypothalamus (VMN) to facilitate lordosis behavior. Estradiol treatment in vivo induces alpha(1b)-adrenoreceptor mRNA and increases the density of alpha(1B)-adrenoreceptor binding in the hypothalamus. Activation of hypothalamic alpha(1)-adrenoceptors also facilitates estrogen-dependent lordosis. To investigate the cellular mechanisms of adrenergic effects on VMN neurons, whole-cell patch-clamp recordings were carried out on hypothalamic slices from control and estradiol-treated female rats. In control slices, bath application of the alpha(1)-agonist phenylephrine (PHE; 10 microM) depolarized 10 of 25 neurons (40%), hyperpolarized three neurons (12%), and had no effect on 12 neurons (48%). The depolarization was associated with decreased membrane conductance, and this current had a reversal potential close to the K(+) equilibrium potential. The alpha(1b)-receptor antagonist chloroethylclonidine (10 microM) blocked the depolarization produced by PHE in all cells. From estradiol-treated rats, significantly more neurons in slices depolarized (71%) and fewer neurons showed no response (17%) to PHE. PHE-induced depolarizations were significantly attenuated with 4-aminopyridine (5 mM) but unaffected by tetraethylammonium chloride (20 mM) or blockers of Na(+) and Ca(2+) channels. These data indicate that alpha(1)-adrenoceptors depolarize VMN neurons by reducing membrane conductance for K(+). Estradiol amplifies alpha(1b)-adrenergic signaling by increasing the proportion of VMN neurons that respond to stimulation of alpha(1b)-adrenergic receptors, which is expected in turn to promote lordosis.


Asunto(s)
Estradiol/farmacología , Hipotálamo/patología , Norepinefrina/farmacología , Fenilefrina/farmacología , Núcleo Hipotalámico Ventromedial/patología , 4-Aminopiridina/química , Animales , Electrofisiología/métodos , Estrógenos/metabolismo , Femenino , Neuronas/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/química , Ratas , Tetraetilamonio/química
9.
J Neurosci ; 29(38): 11965-72, 2009 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-19776282

RESUMEN

Phosphorylation of the NR1 subunit of NMDA receptors (NMDARs) at serine (S) 897 is markedly reduced in schizophrenia patients. However, the role of NR1 S897 phosphorylation in normal synaptic function and adaptive behaviors are unknown. To address these questions, we generated mice in which the NR1 S897 is replaced with alanine (A). This knock-in mutation causes severe impairment in NMDAR synaptic incorporation and NMDAR-mediated synaptic transmission. Furthermore, the phosphomutant animals have reduced AMPA receptor (AMPAR)-mediated synaptic transmission, decreased AMPAR GluR1 subunit in the synapse, and impaired long-term potentiation. Finally, the mutant mice exhibit behavioral deficits in social interaction and sensorimotor gating. Our results suggest that an impairment in NR1 phosphorylation leads to glutamatergic hypofunction that can contribute to behavioral deficits associated with psychiatric disorders.


Asunto(s)
Conducta Animal/fisiología , Plasticidad Neuronal , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Encéfalo/fisiología , Encéfalo/ultraestructura , Técnicas de Sustitución del Gen , Técnicas In Vitro , Potenciación a Largo Plazo/genética , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Plasticidad Neuronal/genética , Neuronas/fisiología , Neuronas/ultraestructura , Fosforilación , Receptores AMPA/metabolismo , Esquizofrenia/genética , Conducta Social , Sinapsis/genética , Sinapsis/fisiología , Sinapsis/ultraestructura , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
10.
Sci Transl Med ; 12(558)2020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32848093

RESUMEN

A major sex difference in Alzheimer's disease (AD) is that men with the disease die earlier than do women. In aging and preclinical AD, men also show more cognitive deficits. Here, we show that the X chromosome affects AD-related vulnerability in mice expressing the human amyloid precursor protein (hAPP), a model of AD. XY-hAPP mice genetically modified to develop testicles or ovaries showed worse mortality and deficits than did XX-hAPP mice with either gonad, indicating a sex chromosome effect. To dissect whether the absence of a second X chromosome or the presence of a Y chromosome conferred a disadvantage on male mice, we varied sex chromosome dosage. With or without a Y chromosome, hAPP mice with one X chromosome showed worse mortality and deficits than did those with two X chromosomes. Thus, adding a second X chromosome conferred resilience to XY males and XO females. In addition, the Y chromosome, its sex-determining region Y gene (Sry), or testicular development modified mortality in hAPP mice with one X chromosome such that XY males with testicles survived longer than did XY or XO females with ovaries. Furthermore, a second X chromosome conferred resilience potentially through the candidate gene Kdm6a, which does not undergo X-linked inactivation. In humans, genetic variation in KDM6A was linked to higher brain expression and associated with less cognitive decline in aging and preclinical AD, suggesting its relevance to human brain health. Our study suggests a potential role for sex chromosomes in modulating disease vulnerability related to AD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/genética , Animales , Femenino , Masculino , Ratones , Caracteres Sexuales , Testículo , Cromosoma X/genética , Cromosoma Y
11.
Eur J Neurosci ; 29(4): 795-801, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19200066

RESUMEN

Estrogens reduce lipocalin-type prostaglandin D synthase (L-PGDS) expression in a region-dependent manner in the mouse preoptic area (POA). This result linked sex hormones with sleep-wake cycle regulation. In turn, the somnogenic effects of prostaglandin D(2) have been shown to be mediated via increases in adenosine, and a select group of sleep-active ventrolateral preoptic nucleus (VLPO) neurons are directly activated by adenosine 2A (A(2A)) agonists. We hypothesized that increased arousal after estrogen administration is mediated by a reduction of L-PGDS and lowered A(2A) receptor expression in the POA. To test this hypothesis, running wheel activity (RWA) of ovariectomized female mice treated with oil or different doses of estradiol benzoate (EB) was studied, followed by quantitative reverse-transcriptase polymerase chain reaction to determine mRNA expression of genes related to sleep and arousal in brain region extracts from oil-treated and EB-treated mice. RWA was increased in estrogen-treated mice, and these effects followed an inverted-U dose-response curve. The most effective dose (1.25 microg EB/capsule) increased RWA more than 2.5-fold, as compared with control animals, and EB doses that were higher or lower were less effective. Increases in RWA were accompanied by decreased L-PGDS mRNA in the POA and decreased A(2A) receptor mRNA in the POA and VLPO. Given that EB-treated animals have higher motor activity and lower levels of L-PGDS and A(2A) receptor mRNAs in sleep-active areas, these correlational findings support the hypothesis that EB may increase behavioral arousal by decreasing the levels of well-known sleep-inducing molecules within the preoptic region.


Asunto(s)
Nivel de Alerta/fisiología , Conducta Animal/fisiología , Encéfalo/metabolismo , Estradiol/análogos & derivados , Expresión Génica , Análisis de Varianza , Animales , Atención , Estradiol/metabolismo , Femenino , Perfilación de la Expresión Génica , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Ratones , Actividad Motora/fisiología , Ovariectomía , Área Preóptica/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Adenosina A2A/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sueño/fisiología , Vigilia/fisiología
12.
Psychoneuroendocrinology ; 33(5): 634-42, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18374493

RESUMEN

Male risk taking and decision making are affected by sex-related cues, with men making poorer and riskier decisions in the presence of females and, or their cues. In non-human species, female cues can also increase male risk taking, reducing their responses to predator threat. As estrogen receptors alpha and beta (ERalpha and ERbeta) are involved in the mediation of social and sexual responses, we investigated their roles in determining the effects of female-associated cues on male risk taking. We examined the effects of brief pre-exposure to the odors of either a novel or familiar estrous female on the avoidance of, and aversive responses to, predator threat (cat odor) in ERalpha and ERbeta wild type (alphaERWT, betaERWT) and gene-deleted (knockout, alphaERKO, betaERKO) male mice. Exposure of alphaERWT and betaERWT males to the odors of a novel, but not a familiar, estrous female mouse resulted in enhanced risk taking with the males displaying reduced avoidance of, and analgesic responses to, cat odor. In contrast, alphaERKO male mice failed to show any changes in risk taking, while betaERKO males, although displaying greater risk taking, did not distinguish between novel and familiar females, displaying similarly reduced avoidance responses to cat odor after exposure to either a novel or familiar female odor. These findings indicate that the gene for ERalpha is associated with the sexual mechanisms (response to estrous female) and the genes for ERbeta and ERalpha with the social (recognition of novel female) mechanisms underlying the effects of female cues on male risk taking.


Asunto(s)
Señales (Psicología) , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Odorantes , Asunción de Riesgos , Conducta Sexual , Animales , Gatos , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Conducta Predatoria , Reconocimiento en Psicología
13.
Neuromuscul Disord ; 27(7): 635-645, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28554556

RESUMEN

Enzyme-linked and electrochemiluminescence immunoassays were developed for quantification of amino (N-) terminal fragments of the skeletal muscle protein titin (N-ter titin) and qualified for use in detection of urinary N-ter titin excretion. Urine from normal subjects contained a small but measurable level of N-ter titin (1.0 ± 0.4 ng/ml). A 365-fold increase (365.4 ± 65.0, P = 0.0001) in urinary N-ter titin excretion was seen in Duchene muscular dystrophy (DMD) patients. Urinary N-ter titin was also evaluated in dystrophin deficient rodent models. Mdx mice exhibited low urinary N-ter titin levels at 2 weeks of age followed by a robust and sustained elevation starting at 3 weeks of age, coincident with the development of systemic skeletal muscle damage in this model; fold elevation could not be determined because urinary N-ter titin was not detected in age-matched wild type mice. Levels of serum creatine kinase and serum skeletal muscle troponin I (TnI) were also low at 2 weeks, elevated at later time points and were significantly correlated with urinary N-ter titin excretion in mdx mice. Corticosteroid treatment of mdx mice resulted in improved exercise performance and lowering of both urinary N-ter titin and serum skeletal muscle TnI concentrations. Low urinary N-ter titin levels were detected in wild type rats (3.0 ± 0.6 ng/ml), while Dmdmdx rats exhibited a 556-fold increase (1652.5 ± 405.7 ng/ml, P = 0.002) (both at 5 months of age). These results suggest that urinary N-ter titin is present at low basal concentrations in normal urine and increases dramatically coincident with muscle damage produced by dystrophin deficiency. Urinary N-ter titin has potential as a facile, non-invasive and translational biomarker for DMD.


Asunto(s)
Conectina/orina , Distrofia Muscular de Duchenne/orina , Adolescente , Corticoesteroides/uso terapéutico , Factores de Edad , Animales , Estudios de Casos y Controles , Niño , Preescolar , Conectina/sangre , Creatina Quinasa/sangre , Estudios Transversales , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos mdx , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular Animal/orina , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/genética
14.
Prog Brain Res ; 158: 243-72, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17027700

RESUMEN

Sex hormone effects on hypothalamic neurons have been worked out to a point where receptor mechanisms are relatively well understood, a neural circuit for a sex steroid-dependent behavior has been determined, and several functional genomic regulations have been discovered and conceptualized. With that knowledge in hand, we approach deeper problems of explaining sexual arousal and generalized CNS arousal. After a brief summary of arousal mechanisms, we focus on three chemical systems which signal generalized arousal and impact hormone-dependent hypothalamic neurons of behavioral importance: histamine, norepinephrine and enkephalin.


Asunto(s)
Hormonas Esteroides Gonadales/fisiología , Hipotálamo/fisiología , Modelos Neurológicos , Sistemas Neurosecretores/fisiología , Conducta Sexual/fisiología , Animales , Sistema Nervioso Central/fisiología , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Genómica , Humanos , Masculino , Neuronas/metabolismo
15.
Physiol Behav ; 88(3): 283-93, 2006 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-16769096

RESUMEN

It now seems possible to move beyond analyzing only the mechanisms for specific sexual behaviors to the analysis of 'generalized arousal' that underlies all motivated behaviors. Our science has advanced sufficiently to attack mechanisms linking specific motivations to these general arousal mechanisms that intrinsically activate all biologically-regulated behaviors including ingestive behaviors. Learning from the well-developed reproductive behavior paradigm, we know that sex hormone effects on hypothalamic neurons have been studied to a point where receptor mechanisms are relatively well understood, a neural circuit for a sex steroid-dependent behavior has been worked out, and several functional genomic regulations have been discovered. Here we focus for the first time on three chemical systems that signal 'generalized arousal' and which impact hormone-dependent hypothalamic neurons of importance to sexual arousal: histamine, norepinephrine and enkephalin. Progress in linking generalized arousal to specific motivational mechanisms is reviewed.


Asunto(s)
Nivel de Alerta/fisiología , Estrógenos/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Receptores Histamínicos/fisiología , Receptores Opioides/fisiología , Conducta Sexual Animal/fisiología , Animales , Sistema Nervioso Central/fisiología , Femenino , Masculino , Postura , Transducción de Señal/fisiología , Activación Transcripcional/fisiología
16.
Nat Commun ; 6: 8897, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26615780

RESUMEN

Maintaining DNA integrity is vital for all cells and organisms. Defective DNA repair may contribute to neurological disorders, including Alzheimer's disease (AD). We found reduced levels of BRCA1, but not of other DNA repair factors, in the brains of AD patients and human amyloid precursor protein (hAPP) transgenic mice. Amyloid-ß oligomers reduced BRCA1 levels in primary neuronal cultures. In wild-type mice, knocking down neuronal BRCA1 in the dentate gyrus caused increased DNA double-strand breaks, neuronal shrinkage, synaptic plasticity impairments, and learning and memory deficits, but not apoptosis. Low levels of hAPP/Amyloid-ß overexpression exacerbated these effects. Physiological neuronal activation increased BRCA1 levels, whereas stimulating predominantly extrasynaptic N-methyl-D-aspartate receptors promoted the proteasomal degradation of BRCA1. We conclude that BRCA1 is regulated by neuronal activity, protects the neuronal genome, and critically supports neuronal integrity and cognitive functions. Pathological accumulation of Aß depletes neuronal BRCA1, which may contribute to cognitive deficits in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Proteína BRCA1/deficiencia , Encéfalo/metabolismo , Reparación del ADN , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Proteína BRCA1/genética , Encéfalo/fisiopatología , Cognición , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo
17.
Nat Neurosci ; 18(3): 423-34, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25622143

RESUMEN

Astrocytes express a variety of G protein-coupled receptors and might influence cognitive functions, such as learning and memory. However, the roles of astrocytic Gs-coupled receptors in cognitive function are not known. We found that humans with Alzheimer's disease (AD) had increased levels of the Gs-coupled adenosine receptor A2A in astrocytes. Conditional genetic removal of these receptors enhanced long-term memory in young and aging mice and increased the levels of Arc (also known as Arg3.1), an immediate-early gene that is required for long-term memory. Chemogenetic activation of astrocytic Gs-coupled signaling reduced long-term memory in mice without affecting learning. Like humans with AD, aging mice expressing human amyloid precursor protein (hAPP) showed increased levels of astrocytic A2A receptors. Conditional genetic removal of these receptors enhanced memory in aging hAPP mice. Together, these findings establish a regulatory role for astrocytic Gs-coupled receptors in memory and suggest that AD-linked increases in astrocytic A2A receptor levels contribute to memory loss.


Asunto(s)
Astrocitos/metabolismo , Regulación de la Expresión Génica/fisiología , Memoria a Largo Plazo/fisiología , Receptor de Adenosina A2A/metabolismo , Receptores de Serotonina 5-HT4/metabolismo , Transducción de Señal/fisiología , Enfermedad de Alzheimer/patología , Animales , Animales Recién Nacidos , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Indoles/farmacología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptor de Adenosina A2A/genética , Receptores de Serotonina 5-HT4/genética , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Antagonistas de la Serotonina/farmacología , Sulfonamidas/farmacología
18.
PLoS One ; 10(5): e0125614, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25933020

RESUMEN

In Alzheimer's disease (AD), an extensive accumulation of extracellular amyloid plaques and intraneuronal tau tangles, along with neuronal loss, is evident in distinct brain regions. Staging of tau pathology by postmortem analysis of AD subjects suggests a sequence of initiation and subsequent spread of neurofibrillary tau tangles along defined brain anatomical pathways. Further, the severity of cognitive deficits correlates with the degree and extent of tau pathology. In this study, we demonstrate that phospho-tau (p-tau) antibodies, PHF6 and PHF13, can prevent the induction of tau pathology in primary neuron cultures. The impact of passive immunotherapy on the formation and spread of tau pathology, as well as functional deficits, was subsequently evaluated with these antibodies in two distinct transgenic mouse tauopathy models. The rTg4510 transgenic mouse is characterized by inducible over-expression of P301L mutant tau, and exhibits robust age-dependent brain tau pathology. Systemic treatment with PHF6 and PHF13 from 3 to 6 months of age led to a significant decline in brain and CSF p-tau levels. In a second model, injection of preformed tau fibrils (PFFs) comprised of recombinant tau protein encompassing the microtubule-repeat domains into the cortex and hippocampus of young P301S mutant tau over-expressing mice (PS19) led to robust tau pathology on the ipsilateral side with evidence of spread to distant sites, including the contralateral hippocampus and bilateral entorhinal cortex 4 weeks post-injection. Systemic treatment with PHF13 led to a significant decline in the spread of tau pathology in this model. The reduction in tau species after p-tau antibody treatment was associated with an improvement in novel-object recognition memory test in both models. These studies provide evidence supporting the use of tau immunotherapy as a potential treatment option for AD and other tauopathies.


Asunto(s)
Enfermedad de Alzheimer/terapia , Anticuerpos Monoclonales/farmacología , Trastornos del Conocimiento/terapia , Inmunización Pasiva , Fosfoproteínas/farmacología , Proteínas tau/antagonistas & inhibidores , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/inmunología , Corteza Cerebral/patología , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/inmunología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Regulación de la Expresión Génica , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Hipocampo/patología , Masculino , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/patología , Cultivo Primario de Células , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Transducción de Señal , Resultado del Tratamiento , Proteínas tau/genética , Proteínas tau/inmunología
19.
PLoS One ; 9(8): e106050, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25153994

RESUMEN

Filamentous inclusions of the microtubule-associated protein, tau, define a variety of neurodegenerative diseases known as tauopathies, including Alzheimer's disease (AD). To better understand the role of tau-mediated effects on pathophysiology and global central nervous system function, we extensively characterized gene expression, pathology and behavior of the rTg4510 mouse model, which overexpresses a mutant form of human tau that causes Frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). We found that the most predominantly altered gene expression pathways in rTg4510 mice were in inflammatory processes. These results closely matched the causal immune function and microglial gene-regulatory network recently identified in AD. We identified additional gene expression changes by laser microdissecting specific regions of the hippocampus, which highlighted alterations in neuronal network activity. Expression of inflammatory genes and markers of neuronal activity changed as a function of age in rTg4510 mice and coincided with behavioral deficits. Inflammatory changes were tau-dependent, as they were reversed by suppression of the tau transgene. Our results suggest that the alterations in microglial phenotypes that appear to contribute to the pathogenesis of Alzheimer's disease may be driven by tau dysfunction, in addition to the direct effects of beta-amyloid.


Asunto(s)
Enfermedad de Alzheimer/genética , Expresión Génica/genética , Redes Reguladoras de Genes/genética , Inflamación/genética , Proteínas tau/genética , Animales , Cromosomas Humanos Par 17/genética , Modelos Animales de Enfermedad , Femenino , Demencia Frontotemporal/genética , Hipocampo/metabolismo , Humanos , Ratones , Microglía/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Enfermedades Neurodegenerativas/genética , Neuronas/metabolismo , Trastornos Parkinsonianos/genética
20.
Neurobiol Aging ; 34(6): 1523-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23332171

RESUMEN

The microtubule-associated protein tau is expressed throughout the nervous system, most highly in neurons but also in glial cells. Its functions in adult and aging mammals remain to be defined. Previous studies in mouse models found either protective or detrimental effects of genetic tau ablation. Though tau ablation prevented synaptic, network, and cognitive dysfunctions in several models of Alzheimer's disease and made mice more resistant to epileptic seizures, a recent study described a parkinsonian phenotype in aging Tau knockout mice. Here we tested cognition and motor functions in Tau(+/+), Tau(+/-), and Tau(-/-) mice at approximately 1 and 2 years of age. Tau ablation did not impair cognition and caused only minor motor deficits that were much more subtle than those associated with the aging process. Tau ablation caused a mild increase in body weight, which correlated with and might have contributed to some of the motor deficits. However, tau ablation did not cause significant dopaminergic impairments, and dopamine treatment did not improve the motor deficits, suggesting that they do not reflect extrapyramidal dysfunction.


Asunto(s)
Envejecimiento/fisiología , Envejecimiento/psicología , Cognición/fisiología , Dopamina , Trastornos de la Destreza Motora/metabolismo , Proteínas tau/deficiencia , Animales , Dopamina/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos de la Destreza Motora/genética , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA