Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 75(1): 13-25.e5, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31151856

RESUMEN

Arc is a synaptic protein essential for memory consolidation. Recent studies indicate that Arc originates in evolution from a Ty3-Gypsy retrotransposon GAG domain. The N-lobe of Arc GAG domain acquired a hydrophobic binding pocket in higher vertebrates that is essential for Arc's canonical function to weaken excitatory synapses. Here, we report that Arc GAG also acquired phosphorylation sites that can acutely regulate its synaptic function. CaMKII phosphorylates the N-lobe of the Arc GAG domain and disrupts an interaction surface essential for high-order oligomerization. In Purkinje neurons, CaMKII phosphorylation acutely reverses Arc's synaptic action. Mutant Arc that cannot be phosphorylated by CaMKII enhances metabotropic receptor-dependent depression in the hippocampus but does not alter baseline synaptic transmission or long-term potentiation. Behavioral studies indicate that hippocampus- and amygdala-dependent learning requires Arc GAG domain phosphorylation. These studies provide an atomic model for dynamic and local control of Arc function underlying synaptic plasticity and memory.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células de Purkinje/metabolismo , Secuencia de Aminoácidos , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/metabolismo , Animales , Sitios de Unión , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/química , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Técnicas de Sustitución del Gen , Células HEK293 , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Fosforilación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Células de Purkinje/citología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Sinapsis/fisiología , Transmisión Sináptica
2.
Cell ; 145(5): 758-72, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21565394

RESUMEN

We have created a mouse genetic model that mimics a human mutation of Shank3 that deletes the C terminus and is associated with autism. Expressed as a single copy [Shank3(+/ΔC) mice], Shank3ΔC protein interacts with the wild-type (WT) gene product and results in >90% reduction of Shank3 at synapses. This "gain-of-function" phenotype is linked to increased polyubiquitination of WT Shank3 and its redistribution into proteasomes. Similarly, the NR1 subunit of the NMDA receptor is reduced at synapses with increased polyubiquitination. Assays of postsynaptic density proteins, spine morphology, and synapse number are unchanged in Shank3(+/ΔC) mice, but the amplitude of NMDAR responses is reduced together with reduced NMDAR-dependent LTP and LTD. Reciprocally, mGluR-dependent LTD is markedly enhanced. Shank3(+/ΔC) mice show behavioral deficits suggestive of autism and reduced NMDA receptor function. These studies reveal a mechanism distinct from haploinsufficiency by which mutations of Shank3 can evoke an autism-like disorder.


Asunto(s)
Trastorno Autístico/genética , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Trastorno Autístico/metabolismo , Trastorno Autístico/fisiopatología , Proteínas Portadoras/genética , Hipocampo/metabolismo , Humanos , Relaciones Interpersonales , Potenciación a Largo Plazo , Depresión Sináptica a Largo Plazo , Ratones , Proteínas de Microfilamentos , Proteínas del Tejido Nervioso , Receptores de Glutamato Metabotrópico/metabolismo , Sinapsis/metabolismo , Ubiquitinación
3.
Behav Brain Funct ; 20(1): 7, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38575965

RESUMEN

BACKGROUND: Alzheimer's disease (AD) and amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) are debilitating neurodegenerative diseases for which there are currently no cures. Familial cases with known genetic causes make up less than 10% of these diseases, and little is known about the underlying mechanisms that contribute to sporadic disease. Accordingly, it is important to expand investigations into possible pathways that may contribute to disease pathophysiology. Glycerophosphodiester phosphodiesterase 2 (GDE2 or GDPD5) is a membrane-bound enzyme that acts at the cell surface to cleave the glycosylphosphatidylinositol (GPI)-anchor that tethers distinct proteins to the membrane. GDE2 abnormally accumulates in intracellular compartments in the brain of patients with AD, ALS, and ALS/FTD, indicative of GDE2 dysfunction. Mice lacking GDE2 (Gde2KO) show neurodegenerative changes such as neuronal loss, reduced synaptic proteins and synapse loss, and increased Aß deposition, raising the possibility that GDE2 disruption in disease might contribute to disease pathophysiology. However, the effect of GDE2 loss on behavioral function and learning/memory has not been characterized. RESULTS: Here, we show that GDE2 is expressed throughout the adult mouse brain in areas including the cortex, hippocampus, habenula, thalamus, and amygdala. Gde2KO and WT mice were tested in a set of behavioral tasks between 7 and 16 months of age. Compared to WT, Gde2KO mice display moderate hyperactivity that becomes more pronounced with age across a variety of behavioral tests assessing novelty-induced exploratory activity. Additionally, Gde2KO mice show reduced startle response, with females showing additional defects in prepulse inhibition. No changes in anxiety-associated behaviors were found, but Gde2KOs show reduced sociability. Notably, aged Gde2KO mice demonstrate impaired short/long-term spatial memory and cued fear memory/secondary contextual fear acquisition. CONCLUSIONS: Taken together, these observations suggest that loss of GDE2 leads to behavioral deficits, some of which are seen in neurodegenerative disease models, implying that loss of GDE2 may be an important contributor to phenotypes associated with neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Enfermedades Neurodegenerativas , Anciano , Animales , Femenino , Humanos , Ratones , Enfermedad de Alzheimer/genética , Esclerosis Amiotrófica Lateral/genética , Demencia Frontotemporal/genética , Memoria , Trastornos de la Memoria/genética , Ratones Transgénicos , Enfermedades Neurodegenerativas/genética
4.
Gen Comp Endocrinol ; 330: 114126, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36122793

RESUMEN

Alzheimer's disease (AD) is a progressive, dementing, whole-body disorder that presents with decline in cognitive, behavioral, and emotional functions, as well as endocrine dysregulation. The etiology of AD is not fully understood but stress- and anxiety-related hormones may play a role in its development and trajectory. The glucocorticoid cascade hypothesis posits that levels of glucocorticoids increase with age, leading to dysregulated negative feedback, further elevated glucocorticoids, and resulting neuropathology. We examined the impact of age (from 2 to 10 months) and stressor exposure (predator odor) on hormone levels (corticosterone and ghrelin), anxiety-like behavior (open field and light dark tests), and memory-related behavior (novel object recognition; NOR), and whether these various measures correlated with neuropathology (hippocampus and cortex amyloid beta, Aß) in male and female APPswe/PS1dE9 transgenic and non-transgenic mice. Additionally, we performed exploratory analyses to probe if the open field and light dark test as commonly used tasks to assess anxiety levels were correlated. Consistent with the glucocorticoid cascade hypothesis, baseline corticosterone increased with age. Predator odor exposure elevated corticosterone at each age, but in contrast to the glucocorticoid cascade hypothesis, the magnitude of stressor-induced elevations in corticosterone levels did not increase with age. Overall, transgenic mice had higher post-stressor, but not baseline, corticosterone than non-transgenic mice, and across both genotypes, females consistently had higher (baseline and post-stressor) corticosterone than males. Behavior in the open field test primarily showed decreased locomotion with age, and this was pronounced in transgenic females. Anxiety-like behaviors in the light dark test were exacerbated following predator odor, and female transgenic mice were the most impacted. Compared to transgenic males, transgenic females had higher Aß concentrations and showed more anxiety-like behavior. Performance on the NOR did not differ significantly between genotypes. Lastly, we did not find robust, statistically significant correlations among corticosterone, ghrelin, recognition memory, anxiety-like behaviors, or Aß, suggesting outcomes are not strongly related on the individual level. Our data suggest that despite Aß accumulation in the hippocampus and cortex, male and female APPswePS1dE9 transgenic mice do not differ robustly from their non-transgenic littermates in physiological, endocrine, and behavioral measures at the range of ages studied here.


Asunto(s)
Enfermedad de Alzheimer , Glucocorticoides , Ratones , Masculino , Femenino , Animales , Corticosterona , Ghrelina , Péptidos beta-Amiloides/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Ansiedad , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/psicología , Ratones Transgénicos , Envejecimiento/fisiología , Estrés Psicológico
5.
Proc Natl Acad Sci U S A ; 116(5): 1686-1691, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30635412

RESUMEN

While neuroinflammation is an evolving concept and the cells involved and their functions are being defined, microglia are understood to be a key cellular mediator of brain injury and repair. The ability to measure microglial activity specifically and noninvasively would be a boon to the study of neuroinflammation, which is involved in a wide variety of neuropsychiatric disorders including traumatic brain injury, demyelinating disease, Alzheimer's disease (AD), and Parkinson's disease, among others. We have developed [11C]CPPC [5-cyano-N-(4-(4-[11C]methylpiperazin-1-yl)-2-(piperidin-1-yl)phenyl)furan-2-carboxamide], a positron-emitting, high-affinity ligand that is specific for the macrophage colony-stimulating factor 1 receptor (CSF1R), the expression of which is essentially restricted to microglia within brain. [11C]CPPC demonstrates high and specific brain uptake in a murine and nonhuman primate lipopolysaccharide model of neuroinflammation. It also shows specific and elevated uptake in a murine model of AD, experimental allergic encephalomyelitis murine model of demyelination and in postmortem brain tissue of patients with AD. Radiation dosimetry in mice indicated [11C]CPPC to be safe for future human studies. [11C]CPPC can be synthesized in sufficient radiochemical yield, purity, and specific radioactivity and possesses binding specificity in relevant models that indicate potential for human PET imaging of CSF1R and the microglial component of neuroinflammation.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/metabolismo , Microglía/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Placa Amiloide/metabolismo , Tomografía de Emisión de Positrones/métodos , Primates , Radiofármacos/metabolismo
6.
Am J Geriatr Psychiatry ; 28(1): 1-19, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31278012

RESUMEN

Psychosis in Alzheimer Disease (AD) represents a distinct clinicopathologic variant associated with increased cognitive and functional morbidity and an accelerated disease course. To date, extant treatments offer modest benefits with significant risks. The development of new pharmacologic treatments for psychosis in AD would be facilitated by validated preclinical models with which to test candidate interventions. The current review provides a brief summary of the process of validating animal models of human disease together with a critical analysis of the challenges posed in attempting to apply those standards to AD-related behavioral models. An overview of phenotypic analogues of human cognitive and behavioral impairments, with an emphasis on those relevant to psychosis, in AD-related mouse models is provided, followed by an update on recent progress in efforts to translate findings in the pathophysiology of psychotic AD into novel models. Finally, some future directions are suggested to expand the catalogue of psychosis-relevant phenotypes that may provide a sturdier framework for model development and targets for preclinical treatment outcomes.


Asunto(s)
Enfermedad de Alzheimer , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Trastornos Psicóticos , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Trastornos Psicóticos/etiología , Trastornos Psicóticos/genética , Trastornos Psicóticos/metabolismo , Trastornos Psicóticos/fisiopatología
7.
Acta Neuropathol ; 138(4): 551-574, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31168644

RESUMEN

Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are clinically and neuropathologically highly related α-synucleinopathies that collectively constitute the second leading cause of neurodegenerative dementias. Genetic and neuropathological studies directly implicate α-synuclein (αS) abnormalities in PDD and DLB pathogenesis. However, it is currently unknown how αS abnormalities contribute to memory loss, particularly since forebrain neuronal loss in PDD and DLB is less severe than in Alzheimer's disease. Previously, we found that familial Parkinson's disease-linked human mutant A53T αS causes aberrant localization of the microtubule-associated protein tau to postsynaptic spines in neurons, leading to postsynaptic deficits. Thus, we directly tested if the synaptic and memory deficits in a mouse model of α-synucleinopathy (TgA53T) are mediated by tau. TgA53T mice exhibit progressive memory deficits associated with postsynaptic deficits in the absence of obvious neuropathological and neurodegenerative changes in the hippocampus. Significantly, removal of endogenous mouse tau expression in TgA53T mice (TgA53T/mTau-/-), achieved by mating TgA53T mice to mouse tau-knockout mice, completely ameliorates cognitive dysfunction and concurrent synaptic deficits without affecting αS expression or accumulation of selected toxic αS oligomers. Among the known tau-dependent effects, memory deficits in TgA53T mice were associated with hippocampal circuit remodeling linked to chronic network hyperexcitability. This remodeling was absent in TgA53T/mTau-/- mice, indicating that postsynaptic deficits, aberrant network hyperactivity, and memory deficits are mechanistically linked. Our results directly implicate tau as a mediator of specific human mutant A53T αS-mediated abnormalities related to deficits in hippocampal neurotransmission and suggest a mechanism for memory impairment that occurs as a consequence of synaptic dysfunction rather than synaptic or neuronal loss. We hypothesize that these initial synaptic deficits contribute to network hyperexcitability which, in turn, exacerbate cognitive dysfunction. Our results indicate that these synaptic changes present potential therapeutic targets for amelioration of memory deficits in α-synucleinopathies.


Asunto(s)
Trastornos de la Memoria/metabolismo , Sinapsis/metabolismo , Sinucleinopatías/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Ratones , Ratones Transgénicos , Plasticidad Neuronal , Neuronas/metabolismo , Neuronas/patología , Sinapsis/patología , Sinucleinopatías/genética , Sinucleinopatías/patología , alfa-Sinucleína/genética , Proteínas tau/genética
8.
Neurobiol Dis ; 105: 33-41, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28511918

RESUMEN

Neuropathological and neuroimaging studies have consistently demonstrated degeneration of monoamine systems, especially the serotonin system, in normal aging and Alzheimer's disease. The evidence for degeneration of the serotonin system in mild cognitive impairment is limited. Thus, the goal of the present study was to measure the serotonin transporter in vivo in mild cognitive impairment and healthy controls. The serotonin transporter is a selective marker of serotonin terminals and of the integrity of serotonin projections to cortical, subcortical and limbic regions and is found in high concentrations in the serotonergic cell bodies of origin of these projections (raphe nuclei). Twenty-eight participants with mild cognitive impairment (age 66.6±6.9, 16 males) and 28 healthy, cognitively normal, demographically matched controls (age 66.2±7.1, 15 males) underwent magnetic resonance imaging for measurement of grey matter volumes and high-resolution positron emission tomography with well-established radiotracers for the serotonin transporter and regional cerebral blood flow. Beta-amyloid imaging was performed to evaluate, in combination with the neuropsychological testing, the likelihood of subsequent cognitive decline in the participants with mild cognitive impairment. The following hypotheses were tested: 1) the serotonin transporter would be lower in mild cognitive impairment compared to controls in cortical and limbic regions, 2) in mild cognitive impairment relative to controls, the serotonin transporter would be lower to a greater extent and observed in a more widespread pattern than lower grey matter volumes or lower regional cerebral blood flow and 3) lower cortical and limbic serotonin transporters would be correlated with greater deficits in auditory-verbal and visual-spatial memory in mild cognitive impairment, not in controls. Reduced serotonin transporter availability was observed in mild cognitive impairment compared to controls in cortical and limbic areas typically affected by Alzheimer's disease pathology, as well as in sensory and motor areas, striatum and thalamus that are relatively spared in Alzheimer's disease. The reduction of the serotonin transporter in mild cognitive impairment was greater than grey matter atrophy or reductions in regional cerebral blood flow compared to controls. Lower cortical serotonin transporters were associated with worse performance on tests of auditory-verbal and visual-spatial memory in mild cognitive impairment, not in controls. The serotonin system may represent an important target for prevention and treatment of MCI, particularly the post-synaptic receptors (5-HT4 and 5-HT6), which may not be as severely affected as presynaptic aspects of the serotonin system, as indicated by the observation of lower serotonin transporters in MCI relative to healthy controls.


Asunto(s)
Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/patología , Imagen Molecular , Degeneración Nerviosa/diagnóstico por imagen , Serotonina/metabolismo , Anciano , Enfermedad de Alzheimer/complicaciones , Bencilaminas/metabolismo , Circulación Cerebrovascular , Femenino , Sustancia Gris , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/fisiopatología , Pruebas Neuropsicológicas , Tomografía de Emisión de Positrones , Proteínas de Transporte de Serotonina en la Membrana Plasmática
9.
Hum Brain Mapp ; 38(7): 3391-3401, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28379618

RESUMEN

Resting-state functional connectivity alterations have been demonstrated in Alzheimer's disease (AD) and mild cognitive impairment (MCI) before the observation of AD neuropathology, but mechanisms driving these changes are not well understood. Serotonin neurodegeneration has been observed in MCI and AD and is associated with cognitive deficits and neuropsychiatric symptoms, but the role of the serotonin system in relation to brain network dysfunction has not been a major focus of investigation. The current study investigated the relationship between serotonin transporter availability (SERT; measured using positron emission tomography) and brain network functional connectivity (measured using resting-state functional MRI) in 20 participants with MCI and 21 healthy controls. Two SERT regions of interest were selected for the analysis: the Dorsal Raphe Nuclei (DRN) and the precuneus which represent the cell bodies of origin and a cortical target of projections of the serotonin system, respectively. Both regions show decreased SERT in MCI compared to controls and are the site of early AD pathology. Average resting-state functional connectivity did not differ between MCI and controls. Decreased SERT in DRN was associated with lower hippocampal resting-state connectivity in MCI participants compared to controls. Decreased SERT in the right precuneus was also associated with lower resting-state connectivity of the retrosplenial cortex to the dorsal lateral prefrontal cortex and higher resting-state connectivity of the retrosplenial cortex to the posterior cingulate and in patients with MCI but not in controls. These results suggest that a serotonergic mechanism may underlie changes in brain functional connectivity in MCI. Hum Brain Mapp 38:3391-3401, 2017. © 2017 Wiley Periodicals, Inc.

10.
Neurobiol Dis ; 96: 171-185, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27569580

RESUMEN

Sex differences are a well-known phenomenon in Alzheimer's disease (AD), with women having a higher risk for AD than men. Many AD mouse models display a similar sex-dependent pattern, with females showing earlier cognitive deficits and more severe neuropathology than males. However, whether those differences are relevant to human disease is unclear. Here we show that in AD mouse models that overexpress amyloid precursor protein (APP) under control of the prion protein promoter (PrP), female transgenic mice have higher APP expression than males, complicating interpretations of the role of sex-related factors in such models. By contrast, in a tTa:APPsi model, in which APP expression is driven by the tetracycline transactivator (tTa) from the CaMKIIα promoter, there are no sex-related differences in expression or processing of APP. In addition, the levels of Aß dimers and tetramers, as well as Aß peptide accumulation, are similar between sexes. Behavioral testing demonstrated that both male and female tTa:APPsi mice develop age-dependent deficits in spatial recognition memory and conditional freezing to context. These cognitive deficits were accompanied by habituation-associated hyperlocomotion and startle hyper-reactivity. Significant sex-related dimorphisms were observed, due to females showing earlier onsets of the deficits in conditioned freezing and hyperlocomotion. In addition, tTa:APPsi males but not females demonstrated a lack of novelty-induced activation. Both males and females showed atrophy of the dentate gyrus (DG) of the dorsal hippocampus, associated with widening of the pyramidal layer of the CA1 area in both sexes. Ventral DG was preserved. Sex-related differences were limited to the DG, with females showing more advanced degeneration than males. Collectively, our data show that the tTa:APPsi model is characterized by a lack of sex-related differences in APP expression, making this model useful in deciphering the mechanisms of sex differences in AD pathogenesis. Sex-related dimorphisms observed in this model under conditions of equal APP expression between sexes suggest a higher sensitivity of females to the effects of APP and/or Aß production.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Giro Dentado/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Animales , Atrofia/etiología , Atrofia/patología , Condicionamiento Psicológico/fisiología , Modelos Animales de Enfermedad , Miedo/fisiología , Femenino , Humanos , Locomoción/genética , Masculino , Ratones , Ratones Transgénicos , Modelos Biológicos , Mutación/genética , Presenilina-1/genética , Reconocimiento en Psicología/fisiología , Factores Sexuales , Tetraciclina/farmacología
11.
J Neurosci ; 33(9): 3765-79, 2013 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-23447589

RESUMEN

Transgenic mice that express mutant amyloid precursor protein (APPsi) using tet-Off vector systems provide an alternative model for assessing short- and long-term effects of Aß-targeting therapies on phenotypes related to the deposition of Alzheimer-type amyloid. Here we use such a model, termed APPsi:tTA, to determine what phenotypes persist in mice with high amyloid burden after new production of APP/Aß has been suppressed. We find that 12- to 13-month-old APPsi:tTA mice are impaired in cognitive tasks that assess short- and long-term memories. Acutely suppressing new APPsi/Aß production produced highly significant improvements in performing short-term spatial memory tasks, which upon continued suppression translated to superior performance in more demanding tasks that assess long-term spatial memory and working memory. Deficits in episodic-like memory and cognitive flexibility, however, were more persistent. Arresting mutant APPsi production caused a rapid decline in the brain levels of soluble APP ectodomains, full-length APP, and APP C-terminal fragments. As expected, amyloid deposits persisted after new APP/Aß production was inhibited, whereas, unexpectedly, we detected persistent pools of solubilizable, relatively mobile, Aß42. Additionally, we observed persistent levels of Aß-immunoreactive entities that were of a size consistent with SDS-resistant oligomeric assemblies. Thus, in this model with significant amyloid pathology, a rapid amelioration of cognitive deficits was observed despite persistent levels of oligomeric Aß assemblies and low, but detectable solubilizable Aß42 peptides. These findings implicate complex relationships between accumulating Aß and activities of APP, soluble APP ectodomains, and/or APP C-terminal fragments in mediating cognitive deficits in this model of amyloidosis.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Amiloidosis/complicaciones , Amiloidosis/patología , Encéfalo/metabolismo , Trastornos del Conocimiento/etiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/dietoterapia , Amiloidosis/genética , Análisis de Varianza , Animales , Encéfalo/patología , Encéfalo/ultraestructura , Trastornos del Conocimiento/dietoterapia , Trastornos del Conocimiento/patología , Discriminación en Psicología/fisiología , Modelos Animales de Enfermedad , Doxiciclina/administración & dosificación , Ensayo de Inmunoadsorción Enzimática , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Fragmentos de Péptidos/metabolismo , Fenotipo , Placa Amiloide/dietoterapia , Placa Amiloide/metabolismo , Placa Amiloide/patología , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética , Reconocimiento en Psicología/efectos de los fármacos , Percepción Espacial , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología , Factores de Tiempo
12.
Front Behav Neurosci ; 17: 1151833, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37250187

RESUMEN

Memory impairment in Alzheimer's disease patients is thought to be associated with the accumulation of amyloid-beta peptides and tau proteins. However, inconsistent reports of cognitive deficits in pre-clinical studies have raised questions about the link between amyloid-beta and cognitive decline. One possible explanation may be that studies reporting memory deficits often involve behavioral assessments that entail a high stress component. In contrast, in tasks without a high stress component transgenic mice do not consistently show declines in memory. The glucocorticoid cascade hypothesis of aging and the vicious cycle of stress framework suggest that stress exacerbates dementia progression by initiating a cycle of hypothalamic-pituitary-adrenal axis activation and subsequent brain deterioration. Using the APPswe/PS1dE9 mouse model of amyloidosis, we assessed whether stressor exposure prior to testing differentially impaired cognitive performance of aged male and female mice. As part of a larger study, mice performed a delayed match-to-position (DMTP) or a 3-choice serial-reaction time (3CSRT) task. Unexpectedly, these mice did not exhibit cognitive declines during aging. Therefore, at 73 and 74 weeks of age, we exposed mice to a predator odor or forced swim stressor prior to testing to determine if stress revealed cognitive deficits. We predicted stressor exposure would decrease performance accuracy more robustly in transgenic vs. non-transgenic mice. Acute stressor exposure increased accuracy in the DMTP task, but not in the 3CSRT task. Our data suggest that acute stressor exposure prior to testing does not impair cognitive performance in APPswe/PS1dE9 mice.

13.
Behav Brain Res ; 438: 114187, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36343696

RESUMEN

Depression is highly prevalent in Alzheimer Disease (AD); however, there is paucity of studies that focus specifically on the assessment of depression-relevant phenotypes in AD mouse models. Conditional doxycycline-dependent transgenic mouse models reproducing amyloidosis (TetOffAPPsi) and/or tau (TetOffTauP301L) pathology starting at middle age (6 months) were used in this study. As AD patients can experience depressive symptoms relatively early in disease, testing was conducted at early, pre-pathology stages of Aß and/or tau accumulation (starting from 45 days of transgenes expression). Tau-related differences were detected in the Novelty Suppressed Feeding task (NSF), whereas APP-related differences were observed predominantly in measures of the Open Field (OF) and Forced Swim tasks (FST). Effects of combined production of Aß and tau were detected in immobility during the 1st half of the Tail Suspension task (TST). These data demonstrate that results from different tasks are difficult to reconcile using task/variable-centered interpretations in which a single task/variable is assigned an ad-hoc meaning relevant to depression. An alternative, concept-oriented, approach is based on multiple variables/tests, with an understanding of their possible inter-dependence and utilization of statistical approaches that handle correlated data sets. The existence of strong correlations within and between some of the tasks supported utilization of factor analyses (FA). FA explained a similar amount of variability across the genotypes (∼80%) and identified two factors stable across genotypes and representing motor activity and anxiety measures in OF. In contrast, variables related to FST, TST, and NSFT did not demonstrate a structure of factor loadings that would support the existence of a single integral factor of "depressive state" measured by these tasks. In addition, factor loadings varied between genotypes, indicating that genotype-specific between-task correlations need to be considered for interpretations of findings in any single task. In general, this study demonstrates that utilization of multiple tasks to characterize behavioral phenotypes, an approach that is finally gaining more widespread adoption, requires a step of data integration across different behavioral tests for appropriate interpretations.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Depresión , Ratones Transgénicos , Modelos Animales de Enfermedad , Fenotipo , Proteínas tau/genética , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo
14.
J Neurotrauma ; 40(11-12): 1197-1215, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36416234

RESUMEN

Therapies are limited for pediatric traumatic brain injury (TBI), especially for the very young who can experience long-term consequences to learning, memory, and social behavior. Animal models of pediatric TBI have yielded mechanistic insights, but demonstration of clinically relevant long-term behavioral and/or cognitive deficits has been challenging. We characterized short- and long-term outcomes in a controlled cortical impact (CCI) model of pediatric TBI using a panel of tests between 2 weeks and ∼4 months after injury. Male rats with CCI at postnatal Day (PND) 10 were compared with three control groups: Naïve, Anesthesia, and Craniotomy. Motor testing (PND 25-33), novel object recognition (NOR; PND 40-50), and multiple tasks in water maze (WM; PND 65-100) were followed by social interaction tests (PND 120-140). Anesthesia rats performed the same as Naïve rats in all tasks. TBI rats, when compared with Naïve controls, had functional impairments across most tests studied. The most sensitive cognitive processes affected by TBI included those that required fast one-trial learning (NOR, WM), flexibility of acquired memory traces (reversals in WM), response strategies (WM), or recognition memory in the setting of reciprocal social interactions. Both TBI and Craniotomy groups demonstrated increased rates of decision making across several WM tasks, suggesting disinhibition of motor responses. When the TBI group was compared with the Craniotomy group, however, deficits were detected in a limited number of outcomes. The latter included learning speed (WM), cognitive flexibility (WM), and social recognition memory. Notably, effects of craniotomy, when compared with Naïve controls, spanned across multiple tasks, and in some tasks, could reach the effect sizes observed in TBI. These results highlight the importance of appropriate control groups in pediatric CCI models. In addition, the study demonstrates the high sensitivity of comprehensive cognitive testing to detect long-term effects of early-age craniotomy and TBI and provides a template for future testing of experimental therapies.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Ratas , Animales , Masculino , Ratas Sprague-Dawley , Grupos Control , Aprendizaje por Laberinto/fisiología , Lesiones Traumáticas del Encéfalo/complicaciones , Cognición , Modelos Animales de Enfermedad
15.
Biol Psychiatry ; 94(9): 706-720, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36796600

RESUMEN

BACKGROUND: Memory deficits are central to many neuropsychiatric diseases. During acquisition of new information, memories can become vulnerable to interference, yet mechanisms that underlie interference are unknown. METHODS: We describe a novel transduction pathway that links the NMDA receptor (NMDAR) to AKT signaling via the immediate early gene Arc and evaluate its role in memory. The signaling pathway is validated using biochemical tools and transgenic mice, and function is evaluated in assays of synaptic plasticity and behavior. The translational relevance is evaluated in human postmortem brain. RESULTS: Arc is dynamically phosphorylated by CaMKII (calcium/calmodulin-dependent protein kinase II) and binds the NMDAR subunits NR2A/NR2B and a previously unstudied PI3K (phosphoinositide 3-kinase) adapter p55PIK (PIK3R3) in vivo in response to novelty or tetanic stimulation in acute slices. NMDAR-Arc-p55PIK recruits p110α PI3K and mTORC2 (mechanistic target of rapamycin complex 2) to activate AKT. NMDAR-Arc-p55PIK-PI3K-mTORC2-AKT assembly occurs within minutes of exploratory behavior and localizes to sparse synapses throughout hippocampal and cortical regions. Studies using conditional (Nestin-Cre) p55PIK deletion mice indicate that NMDAR-Arc-p55PIK-PI3K-mTORC2-AKT functions to inhibit GSK3 and mediates input-specific metaplasticity that protects potentiated synapses from subsequent depotentiation. p55PIK conditional knockout mice perform normally in multiple behaviors including working memory and long-term memory tasks but exhibit deficits indicative of increased vulnerability to interference in both short-term and long-term paradigms. The NMDAR-AKT transduction complex is reduced in postmortem brain of individuals with early Alzheimer's disease. CONCLUSIONS: A novel function of Arc mediates synapse-specific NMDAR-AKT signaling and metaplasticity that contributes to memory updating and is disrupted in human cognitive disease.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Humanos , Animales , Enfermedad de Alzheimer/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , N-Metilaspartato/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Transducción de Señal , Hipocampo/metabolismo , Ratones Transgénicos , Ratones Noqueados , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo
16.
Neurobiol Aging ; 128: 85-99, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37120419

RESUMEN

Preclinical models of Alzheimer's disease (AD)-related cognitive decline can be useful for developing therapeutics. The current study longitudinally assessed short-term memory, using a delayed matching-to-position (DMTP) task, and attention, using a 3-choice serial reaction time (3CSRT) task, from approximately 18 weeks of age through death or 72 weeks of age in APPswe/PS1dE9 mice, a widely used mouse model of AD-related amyloidosis. Both transgenic (Tg) and non-Tg mice exhibited improvements in DMTP accuracy over time. Breaks in testing reduced DMTP accuracy but accuracy values quickly recovered in both Tg and non-Tg mice. Both Tg and non-Tg mice exhibited high accuracy in the 3CSRT task with breaks in testing briefly reducing accuracy values equivalently in the 2 genotypes. The current results raise the possibility that deficits in Tg APPswe/PS1dE9 mice involve impairments in learning rather than declines in established performances. A better understanding of the factors that determine whether deficits develop will be useful for designing evaluations of potential pharmacotherapeutics and may reveal interventions for clinical application.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Ratones , Animales , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/genética , Ratones Transgénicos , Cognición , Modelos Animales de Enfermedad , Precursor de Proteína beta-Amiloide/genética , Presenilina-1/genética
17.
bioRxiv ; 2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37808783

RESUMEN

Sleep and circadian rhythm disruption (SCRD) is commonly observed in aging, especially in individuals who experience progressive cognitive decline to mild cognitive impairment (MCI) and Alzheimer's disease (AD). However, precise molecular mechanisms underlying the association between SCRD and aging are not fully understood. Orexin A is a well-characterized "sleep neuropeptide" that is expressed in hypothalamic neurons and evokes wake behavior. The importance of Orexin is exemplified in narcolepsy where it is profoundly down-regulated. Interestingly, the synaptic immediate early gene NPTX2 is co-expressed in Orexin neurons and is similarly reduced in narcolepsy. NPTX2 is also down-regulated in CSF of some cognitively normal older individuals and predicts the time of transition from normal cognition to MCI. The association between Orexin and NPTX2 is further evinced here where we observe that Orexin A and NPTX2 are highly correlated in CSF of cognitively normal aged individuals and raises the question of whether SCRD that are typically attributed to Orexin A loss of function may be modified by concomitant NPTX2 down-regulation. Is NPTX2 an effector of sleep or simply a reporter of orexin-dependent SCRD? To address this question, we examined NPTX2 KO mice and found they retain Orexin expression in the brain and so provide an opportunity to examine the specific contribution of NPTX2 to SCRD. Our results reveal that NPTX2 KO mice exhibit a disrupted circadian onset time, coupled with increased activity during the sleep phase, suggesting difficulties in maintaining states. Sleep EEG indicates distinct temporal allocation shifts across vigilance states, characterized by reduced wake and increased NREM time. Evident sleep fragmentation manifests through alterations of event occurrences during Wake and NREM, notably during light transition periods, in conjunction with an increased frequency of sleep transitions in NPTX2 KO mice, particularly between Wake and NREM. EEG spectral analysis indicated significant shifts in power across various frequency bands in the wake, NREM, and REM states, suggestive of disrupted neuronal synchronicity. An intriguing observation is the diminished occurrence of sleep spindles, one of the earliest measures of human sleep disruption, in NPTX2 KO mice. These findings highlight the effector role of NPTX2 loss of function as an instigator of SCRD and a potential mediator of sleep disruption in aging.

18.
Neuroimage Clin ; 37: 103322, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36680976

RESUMEN

BACKGROUND: Degeneration of the serotonin system has been observed in Alzheimer's disease (AD) and in mild cognitive impairment (MCI). In transgenic amyloid mouse models, serotonin degeneration is detected prior to widespread cortical beta-amyloid (Aß) deposition, also suggesting that serotonin degeneration may be observed in preclinical AD. METHODS: The differences in the distribution of serotonin degeneration (reflected by the loss of the serotonin transporter, 5-HTT) relative to Aß deposition was measured with positron emission tomography in a group of individuals with MCI and a group of healthy older adults. A multi-modal partial least squares (mmPLS) algorithm was applied to identify the spatial covariance pattern between 5-HTT availability and Aß deposition. RESULTS: Forty-five individuals with MCI and 35 healthy older adults were studied, 22 and 27 of whom were included in the analyses who were "amyloid positive" and "amyloid negative", respectively. A pattern of lower cortical, subcortical and limbic 5-HTT availability and higher cortical Aß deposition distinguished the MCI from the healthy older control participants. Greater expression of this pattern was correlated with greater deficits in memory and executive function in the MCI group, not in the control group. CONCLUSION: A spatial covariance pattern of lower 5-HTT availability and Aß deposition was observed to a greater extent in an MCI group relative to a control group and was associated with cognitive impairment in the MCI group. The results support the application of mmPLS to understand the neurochemical changes associated with Aß deposition in the course of preclinical AD.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Animales , Ratones , Serotonina , Disfunción Cognitiva/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Tomografía de Emisión de Positrones/métodos , Imagen Molecular
19.
J Alzheimers Dis ; 96(1): 215-227, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37718818

RESUMEN

BACKGROUND: Neuropathological and neuroimaging studies have demonstrated degeneration of the serotonin system in Alzheimer's disease (AD). Neuroimaging studies have extended these observations to the preclinical stages of AD, mild cognitive impairment (MCI). Serotonin degeneration has been observed also in transgenic amyloid mouse models, prior to widespread cortical distribution of amyloid-ß (Aß). OBJECTIVE: The present study evaluated the regional distribution of the serotonin transporter (5-HTT) and of Aß in individuals with MCI and healthy older controls, as well as the contribution of 5-HTT and Aß to cognitive deficits. METHODS: Forty-nine MCI participants and 45 healthy older controls underwent positron emission tomography (PET) imaging of 5-HTT and Aß, structural magnetic resonance imaging and neuropsychological assessments. RESULTS: Lower cortical, striatal, and limbic 5-HTT and higher cortical Aß was observed in MCIs relative to healthy controls. Lower 5-HTT, mainly in limbic regions, was correlated with greater deficits in auditory-verbal and visual-spatial memory and semantic, not phonemic fluency. Higher cortical A ß was associated with greater deficits in auditory-verbal and visual-spatial memory and in semantic, not phonemic fluency. When modeling the association between cognition, gray matter volumes and Aß, inclusion of 5-HTT in limbic and in select cortical regions significantly improved model fit for auditory-verbal and visual-spatial memory and semantic, but not phonemic fluency. CONCLUSIONS: These results support the role of serotonin degeneration in the memory and semantic fluency deficits observed in MCI.


Asunto(s)
Enfermedad de Alzheimer , Trastornos del Conocimiento , Disfunción Cognitiva , Animales , Ratones , Humanos , Serotonina , Disfunción Cognitiva/patología , Trastornos del Conocimiento/complicaciones , Péptidos beta-Amiloides , Enfermedad de Alzheimer/patología , Cognición , Tomografía de Emisión de Positrones
20.
Neurosci Biobehav Rev ; 137: 104660, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35405182

RESUMEN

Toxoplasma exposure can elicit cellular and humoral immune responses. In the case of chronic Toxoplasma infection, these immune responses are long-lasting. Some studies suggest that pre-existing immunity from Toxoplasma infection can shape immune responses and resistance to other pathogens and brain insults later in life. Much evidence has been generated suggesting Toxoplasma infection may contribute to cognitive impairment in the elderly. However, there have also been studies that disagree with the conclusion. Toxoplasma has many strain types, with virulence being the most notable difference. There is also considerable variation in the outcomes following Toxoplasma exposure ranging from resolved to persistent infection. Therefore, the brain microenvironment, particularly cellular constituents, differs based on the infecting strain (virulent versus hypovirulent) and infection stage (resolved versus persistent). Such difference might play a critical role in determining the outcome of the host on subsequent challengings to the brain. The ability of Toxoplasma strains to set up distinct stages for neurodegenerative pathology through varying degrees of virulence provides unique experimental tools for characterizing these pathways.


Asunto(s)
Enfermedad de Alzheimer , Toxoplasma , Anciano , Cognición , Humanos , Inmunidad , Toxoplasma/fisiología , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA