Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
bioRxiv ; 2023 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-37398301

RESUMEN

CRISPR-based genetic screening directly in mammalian tissues in vivo is challenging due to the need for scalable, cell-type selective delivery and recovery of guide RNA libraries. We developed an in vivo adeno-associated virus-based and Cre recombinase-dependent workflow for cell type-selective CRISPR interference screening in mouse tissues. We demonstrate the power of this approach by identifying neuron-essential genes in the mouse brain using a library targeting over 2000 genes.

2.
bioRxiv ; 2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37333294

RESUMEN

Progress in understanding long COVID and developing effective therapeutics is hampered in part by the lack of suitable animal models. Here we used ACE2-transgenic mice recovered from Omicron (BA.1) infection to test for pulmonary and behavioral post-acute sequelae. Through in-depth phenotyping by CyTOF, we demonstrate that naïve mice experiencing a first Omicron infection exhibit profound immune perturbations in the lung after resolving acute infection. This is not observed if mice were first vaccinated with spike-encoding mRNA. The protective effects of vaccination against post-acute sequelae were associated with a highly polyfunctional SARS-CoV-2-specific T cell response that was recalled upon BA.1 breakthrough infection but not seen with BA.1 infection alone. Without vaccination, the chemokine receptor CXCR4 was uniquely upregulated on multiple pulmonary immune subsets in the BA.1 convalescent mice, a process previously connected to severe COVID-19. Taking advantage of recent developments in machine learning and computer vision, we demonstrate that BA.1 convalescent mice exhibited spontaneous behavioral changes, emotional alterations, and cognitive-related deficits in context habituation. Collectively, our data identify immunological and behavioral post-acute sequelae after Omicron infection and uncover a protective effect of vaccination against post-acute pulmonary immune perturbations.

3.
Commun Biol ; 5(1): 1267, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36400882

RESUMEN

Quantification and detection of the hierarchical organization of behavior is a major challenge in neuroscience. Recent advances in markerless pose estimation enable the visualization of high-dimensional spatiotemporal behavioral dynamics of animal motion. However, robust and reliable technical approaches are needed to uncover underlying structure in these data and to segment behavior into discrete hierarchically organized motifs. Here, we present an unsupervised probabilistic deep learning framework that identifies behavioral structure from deep variational embeddings of animal motion (VAME). By using a mouse model of beta amyloidosis as a use case, we show that VAME not only identifies discrete behavioral motifs, but also captures a hierarchical representation of the motif's usage. The approach allows for the grouping of motifs into communities and the detection of differences in community-specific motif usage of individual mouse cohorts that were undetectable by human visual observation. Thus, we present a robust approach for the segmentation of animal motion that is applicable to a wide range of experimental setups, models and conditions without requiring supervised or a-priori human interference.


Asunto(s)
Conducta Animal , Neurociencias , Animales , Humanos , Movimiento (Física)
4.
Nature ; 612(7939): 218-220, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36450951
5.
Mol Neurodegener ; 17(1): 41, 2022 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690868

RESUMEN

BACKGROUND: Genetic mutations underlying familial Alzheimer's disease (AD) were identified decades ago, but the field is still in search of transformative therapies for patients. While mouse models based on overexpression of mutated transgenes have yielded key insights in mechanisms of disease, those models are subject to artifacts, including random genetic integration of the transgene, ectopic expression and non-physiological protein levels. The genetic engineering of novel mouse models using knock-in approaches addresses some of those limitations. With mounting evidence of the role played by microglia in AD, high-dimensional approaches to phenotype microglia in those models are critical to refine our understanding of the immune response in the brain. METHODS: We engineered a novel App knock-in mouse model (AppSAA) using homologous recombination to introduce three disease-causing coding mutations (Swedish, Arctic and Austrian) to the mouse App gene. Amyloid-ß pathology, neurodegeneration, glial responses, brain metabolism and behavioral phenotypes were characterized in heterozygous and homozygous AppSAA mice at different ages in brain and/ or biofluids. Wild type littermate mice were used as experimental controls. We used in situ imaging technologies to define the whole-brain distribution of amyloid plaques and compare it to other AD mouse models and human brain pathology. To further explore the microglial response to AD relevant pathology, we isolated microglia with fibrillar Aß content from the brain and performed transcriptomics and metabolomics analyses and in vivo brain imaging to measure energy metabolism and microglial response. Finally, we also characterized the mice in various behavioral assays. RESULTS: Leveraging multi-omics approaches, we discovered profound alteration of diverse lipids and metabolites as well as an exacerbated disease-associated transcriptomic response in microglia with high intracellular Aß content. The AppSAA knock-in mouse model recapitulates key pathological features of AD such as a progressive accumulation of parenchymal amyloid plaques and vascular amyloid deposits, altered astroglial and microglial responses and elevation of CSF markers of neurodegeneration. Those observations were associated with increased TSPO and FDG-PET brain signals and a hyperactivity phenotype as the animals aged. DISCUSSION: Our findings demonstrate that fibrillar Aß in microglia is associated with lipid dyshomeostasis consistent with lysosomal dysfunction and foam cell phenotypes as well as profound immuno-metabolic perturbations, opening new avenues to further investigate metabolic pathways at play in microglia responding to AD-relevant pathogenesis. The in-depth characterization of pathological hallmarks of AD in this novel and open-access mouse model should serve as a resource for the scientific community to investigate disease-relevant biology.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Microglía/metabolismo , Placa Amiloide/patología , Receptores de GABA/metabolismo
6.
Nat Neurosci ; 24(1): 19-23, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33318667

RESUMEN

Microglial surveillance is a key feature of brain physiology and disease. Here, we found that Gi-dependent microglial dynamics prevent neuronal network hyperexcitability. By generating MgPTX mice to genetically inhibit Gi in microglia, we show that sustained reduction of microglia brain surveillance and directed process motility induced spontaneous seizures and increased hypersynchrony after physiologically evoked neuronal activity in awake adult mice. Thus, Gi-dependent microglia dynamics may prevent hyperexcitability in neurological diseases.


Asunto(s)
Quinasa 1 del Receptor Acoplado a Proteína-G/fisiología , Microglía/fisiología , Red Nerviosa/fisiología , Animales , Señalización del Calcio , Movimiento Celular , Convulsivantes , Electroencefalografía , Vigilancia Inmunológica , Ratones , Microglía/enzimología , Microglía/ultraestructura , Enfermedades del Sistema Nervioso/fisiopatología , Fenómenos Fisiológicos del Sistema Nervioso , Pilocarpina , Convulsiones/fisiopatología , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
7.
Mol Neurodegener ; 15(1): 53, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32921309

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most frequent and costly neurodegenerative disorder. Although diverse lines of evidence suggest that the amyloid precursor protein (APP) is involved in its causation, the precise mechanisms remain unknown and no treatments are available to prevent or halt the disease. A favorite hypothesis has been that APP contributes to AD pathogenesis through the cerebral accumulation of the amyloid-ß peptide (Aß), which is derived from APP through sequential proteolytic cleavage by BACE1 and γ-secretase. However, inhibitors of these enzymes have failed in clinical trials despite clear evidence for target engagement. METHODS: To further elucidate the roles of APP and its metabolites in AD pathogenesis, we analyzed transgenic mice overexpressing wildtype human APP (hAPP) or hAPP carrying mutations that cause autosomal dominant familial AD (FAD), as well as App knock-in mice that do not overexpress hAPP but have two mouse App alleles with FAD mutations and a humanized Aß sequence. RESULTS: Although these lines of mice had marked differences in cortical and hippocampal levels of APP, APP C-terminal fragments, soluble Aß, Aß oligomers and age-dependent amyloid deposition, they all developed cognitive deficits as well as non-convulsive epileptiform activity, a type of network dysfunction that also occurs in a substantive proportion of humans with AD. Pharmacological inhibition of BACE1 effectively reduced levels of amyloidogenic APP C-terminal fragments (C99), soluble Aß, Aß oligomers, and amyloid deposits in transgenic mice expressing FAD-mutant hAPP, but did not improve their network dysfunction and behavioral abnormalities, even when initiated at early stages before amyloid deposits were detectable. CONCLUSIONS: hAPP transgenic and App knock-in mice develop similar pathophysiological alterations. APP and its metabolites contribute to AD-related functional alterations through complex combinatorial mechanisms that may be difficult to block with BACE inhibitors and, possibly, also with other anti-Aß treatments.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal/fisiología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Red Nerviosa/metabolismo , Red Nerviosa/patología
8.
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
9.
Cell Rep ; 30(2): 381-396.e4, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31940483

RESUMEN

NMDA receptors (NMDARs) play subunit-specific roles in synaptic function and are implicated in neuropsychiatric and neurodegenerative disorders. However, the in vivo consequences and therapeutic potential of pharmacologically enhancing NMDAR function via allosteric modulation are largely unknown. We examine the in vivo effects of GNE-0723, a positive allosteric modulator of GluN2A-subunit-containing NMDARs, on brain network and cognitive functions in mouse models of Dravet syndrome (DS) and Alzheimer's disease (AD). GNE-0723 use dependently potentiates synaptic NMDA receptor currents and reduces brain oscillation power with a predominant effect on low-frequency (12-20 Hz) oscillations. Interestingly, DS and AD mouse models display aberrant low-frequency oscillatory power that is tightly correlated with network hypersynchrony. GNE-0723 treatment reduces aberrant low-frequency oscillations and epileptiform discharges and improves cognitive functions in DS and AD mouse models. GluN2A-subunit-containing NMDAR enhancers may have therapeutic benefits in brain disorders with network hypersynchrony and cognitive impairments.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Ciclopropanos/farmacología , Epilepsias Mioclónicas/tratamiento farmacológico , Nitrilos/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Tiazoles/farmacología , Regulación Alostérica/efectos de los fármacos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Pirazoles/farmacología , Receptores de N-Metil-D-Aspartato/agonistas
10.
Neurobiol Aging ; 85: 58-73, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31739167

RESUMEN

Electrophysiology provides a real-time readout of neural functions and network capability in different brain states, on temporal (fractions of milliseconds) and spatial (micro, meso, and macro) scales unmet by other methodologies. However, current international guidelines do not endorse the use of electroencephalographic (EEG)/magnetoencephalographic (MEG) biomarkers in clinical trials performed in patients with Alzheimer's disease (AD), despite a surge in recent validated evidence. This position paper of the ISTAART Electrophysiology Professional Interest Area endorses consolidated and translational electrophysiological techniques applied to both experimental animal models of AD and patients, to probe the effects of AD neuropathology (i.e., brain amyloidosis, tauopathy, and neurodegeneration) on neurophysiological mechanisms underpinning neural excitation/inhibition and neurotransmission as well as brain network dynamics, synchronization, and functional connectivity, reflecting thalamocortical and corticocortical residual capacity. Converging evidence shows relationships between abnormalities in EEG/MEG markers and cognitive deficits in groups of AD patients at different disease stages. The supporting evidence for the application of electrophysiology in AD clinical research as well as drug discovery pathways warrants an international initiative to include the use of EEG/MEG biomarkers in the main multicentric projects planned in AD patients, to produce conclusive findings challenging the present regulatory requirements and guidelines for AD studies.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/fisiopatología , Encéfalo/fisiopatología , Electrofisiología/métodos , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Descubrimiento de Drogas , Electroencefalografía , Potenciales Evocados , Humanos , Magnetoencefalografía
11.
Neuron ; 98(1): 75-89.e5, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29551491

RESUMEN

Inhibitory interneurons regulate the oscillatory rhythms and network synchrony that are required for cognitive functions and disrupted in Alzheimer's disease (AD). Network dysrhythmias in AD and multiple neuropsychiatric disorders are associated with hypofunction of Nav1.1, a voltage-gated sodium channel subunit predominantly expressed in interneurons. We show that Nav1.1-overexpressing, but not wild-type, interneuron transplants derived from the embryonic medial ganglionic eminence (MGE) enhance behavior-dependent gamma oscillatory activity, reduce network hypersynchrony, and improve cognitive functions in human amyloid precursor protein (hAPP)-transgenic mice, which simulate key aspects of AD. Increased Nav1.1 levels accelerated action potential kinetics of transplanted fast-spiking and non-fast-spiking interneurons. Nav1.1-deficient interneuron transplants were sufficient to cause behavioral abnormalities in wild-type mice. We conclude that the efficacy of interneuron transplantation and the function of transplanted cells in an AD-relevant context depend on their Nav1.1 levels. Disease-specific molecular optimization of cell transplants may be required to ensure therapeutic benefits in different conditions.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Ondas Encefálicas/fisiología , Encéfalo/metabolismo , Cognición/fisiología , Interneuronas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/biosíntesis , Potenciales de Acción/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/terapia , Animales , Encéfalo/cirugía , Modelos Animales de Enfermedad , Expresión Génica , Hipocampo/metabolismo , Hipocampo/cirugía , Humanos , Interneuronas/trasplante , Locomoción/fisiología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.1/genética
12.
eNeuro ; 5(6)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30627643

RESUMEN

Alzheimer's disease (AD) begins several decades before the onset of clinical symptoms, at a time when women may still undergo reproductive cycling. Whether ovarian functions alter substrates of AD pathogenesis is unknown. Here we show that ovarian cycle stages significantly modulate AD-related alterations in neural network patterns, cognitive impairments, and pathogenic protein production in the hAPP-J20 mouse model of AD. Female hAPP mice spent more time in estrogen-dominant cycle stages and these ovarian stages worsened AD-related network dysfunction and cognitive impairments. In contrast, progesterone-dominant stages and gonadectomy attenuated these AD-related deficits. Further studies revealed a direct role for estradiol in stimulating neural network excitability and susceptibility to seizures in hAPP mice and increasing amyloid beta levels. Understanding dynamic effects of the ovarian cycle on the female nervous system in disease, including AD, is of critical importance and may differ from effects on a healthy brain. The pattern of ovarian cycle effects on disease-related networks, cognition, and pathogenic protein expression may be relevant to young women at risk for AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Ondas Encefálicas/fisiología , Encéfalo/patología , Trastornos del Conocimiento , Ciclo Menstrual/fisiología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Castración , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Estradiol/metabolismo , Conducta Exploratoria/fisiología , Femenino , Humanos , Ciclo Menstrual/genética , Ratones , Ratones Transgénicos , Mutación/genética , Pentilenotetrazol/toxicidad , Progesterona/metabolismo , Convulsiones/inducido químicamente , Convulsiones/fisiopatología
13.
Nat Rev Neurosci ; 17(12): 777-792, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27829687

RESUMEN

The function of neural circuits and networks can be controlled, in part, by modulating the synchrony of their components' activities. Network hypersynchrony and altered oscillatory rhythmic activity may contribute to cognitive abnormalities in Alzheimer disease (AD). In this condition, network activities that support cognition are altered decades before clinical disease onset, and these alterations predict future pathology and brain atrophy. Although the precise causes and pathophysiological consequences of these network alterations remain to be defined, interneuron dysfunction and network abnormalities have emerged as potential mechanisms of cognitive dysfunction in AD and related disorders. Here, we explore the concept that modulating these mechanisms may help to improve brain function in these conditions.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Encéfalo/fisiopatología , Interneuronas/fisiología , Red Nerviosa/fisiopatología , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Humanos , Interneuronas/patología , Red Nerviosa/patología , Vías Nerviosas/patología , Vías Nerviosas/fisiopatología
14.
Nat Neurosci ; 18(8): 1077-80, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26120963

RESUMEN

Astrocytes modulate neuronal activity and inhibit regeneration. We show that cleaved p75 neurotrophin receptor (p75(NTR)) is a component of the nuclear pore complex (NPC) required for glial scar formation and reduced gamma oscillations in mice via regulation of transforming growth factor (TGF)-ß signaling. Cleaved p75(NTR) interacts with nucleoporins to promote Smad2 nucleocytoplasmic shuttling. Thus, NPC remodeling by regulated intramembrane cleavage of p75(NTR) controls astrocyte-neuronal communication in response to profibrotic factors.


Asunto(s)
Astrocitos/metabolismo , Ritmo Gamma/fisiología , Actividad Motora/fisiología , Poro Nuclear/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Conducta Animal/fisiología , Electroencefalografía , Gliosis/metabolismo , Células HEK293 , Humanos , Hidrocefalia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Proteínas de Complejo Poro Nuclear/metabolismo , Receptor de Factor de Crecimiento Nervioso/deficiencia , Proteína Smad2/metabolismo
15.
J Clin Invest ; 123(6): 2719-29, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23676464

RESUMEN

Adult-onset autosomal-dominant leukodystrophy (ADLD) is a progressive and fatal neurological disorder characterized by early autonomic dysfunction, cognitive impairment, pyramidal tract and cerebellar dysfunction, and white matter loss in the central nervous system. ADLD is caused by duplication of the LMNB1 gene, which results in increased lamin B1 transcripts and protein expression. How duplication of LMNB1 leads to myelin defects is unknown. To address this question, we developed a mouse model of ADLD that overexpresses lamin B1. These mice exhibited cognitive impairment and epilepsy, followed by age-dependent motor deficits. Selective overexpression of lamin B1 in oligodendrocytes also resulted in marked motor deficits and myelin defects, suggesting these deficits are cell autonomous. Proteomic and genome-wide transcriptome studies indicated that lamin B1 overexpression is associated with downregulation of proteolipid protein, a highly abundant myelin sheath component that was previously linked to another myelin-related disorder, Pelizaeus-Merzbacher disease. Furthermore, we found that lamin B1 overexpression leads to reduced occupancy of Yin Yang 1 transcription factor at the promoter region of proteolipid protein. These studies identify a mechanism by which lamin B1 overexpression mediates oligodendrocyte cell-autonomous neuropathology in ADLD and implicate lamin B1 as an important regulator of myelin formation and maintenance during aging.


Asunto(s)
Lamina Tipo B/metabolismo , Oligodendroglía/patología , Enfermedad de Pelizaeus-Merzbacher/metabolismo , Animales , Axones/metabolismo , Axones/patología , Modelos Animales de Enfermedad , Ataxia de la Marcha/metabolismo , Ataxia de la Marcha/patología , Ataxia de la Marcha/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Lamina Tipo B/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora , Proteína Proteolipídica de la Mielina/genética , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Oligodendroglía/metabolismo , Enfermedad de Pelizaeus-Merzbacher/patología , Enfermedad de Pelizaeus-Merzbacher/fisiopatología , Regiones Promotoras Genéticas , Unión Proteica , Prueba de Desempeño de Rotación con Aceleración Constante , Convulsiones/metabolismo , Convulsiones/patología , Convulsiones/fisiopatología , Factor de Transcripción YY1/metabolismo
16.
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
17.
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
18.
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
19.
Methods Mol Biol ; 670: 207-30, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20967593

RESUMEN

RNA in situ hybridization is a powerful technique for examining gene expression in specific cell populations. This method is particularly useful in the central nervous system with its high cellular diversity and dynamic gene expression regulation associated with development, plasticity, neuronal activity, aging, and disease. Standard quantitative techniques such as Western blotting and real-time PCR allow the detection of altered gene or protein expression but provide no information about their cellular source or possible alterations in expression patterns. Here, we describe a step-by-step RNA in situ hybridization method on adult and embryonic brain sections for quantitative neuroscience. We include fully detailed protocols for RNase-free material preparation, perfusion, fixation, sectioning, selection of expressed sequence tag cDNA clones, linearization of cDNA, synthesis of digoxigenin-labeled RNA probes (riboprobes), in situ hybridization on floating and mounted sections, nonradioactive immunohistochemical detection of riboprobes for light and fluorescence microscopy, and double labeling. We also include useful information about quality-control steps, key online sites, commercially available products, stock solutions, and storage. Finally, we provide examples of the utility of this approach in understanding the neuropathogenesis of Alzheimer's disease. With virtually all genomic coding sequences cloned or being cloned into cDNA plasmids, this technique has become highly accessible to explore gene expression profiles at the cellular and brain region level.


Asunto(s)
Encéfalo/metabolismo , Hibridación Fluorescente in Situ/métodos , Animales , Inmunohistoquímica , Microscopía Fluorescente , Modelos Genéticos
20.
Methods Mol Biol ; 670: 245-62, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20967595

RESUMEN

High levels of Aß impair neuronal function at least in part by disrupting normal synaptic transmission and causing dysfunction of neural networks. This network dysfunction includes abnormal synchronization of neuronal activity resulting in epileptiform activity. Over time, this aberrant network activity can lead to the depletion of calcium-dependent proteins, such as calbindin, Fos, and Arc, and compensatory inhibitory remodeling of hippocampal circuits, including GABAergic sprouting and ectopic expression of the inhibitory neuropeptide Y (NPY) in dentate granule cells. Here we present detailed protocols for detecting and quantifying these alterations in mouse models of Alzheimer's disease (AD) by immunohistochemistry. These methods are useful as surrogate measures for detecting chronic aberrant network activity in models of AD and epilepsy. In addition, since we have found that the severity of these changes relates to the degree of Aß-dependent cognitive impairments, the protocols are useful for quantifying biomarkers of cognitive impairment in mouse models of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Trastornos del Conocimiento/metabolismo , Hipocampo/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Biomarcadores/metabolismo , Humanos , Inmunohistoquímica , Técnicas In Vitro , Ratones , Ratones Transgénicos , Neuropéptido Y/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...