Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Hippocampus ; 34(2): 52-57, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38189522

RESUMEN

The famous amnesic patient Henry Molaison (H.M.) died on December 2, 2008. After extensive in situ magnetic resonance imaging in Boston, his brain was removed at autopsy and transported to the University of California San Diego. There the brain was prepared for frozen sectioning and cut into 2401, 70 µm coronal slices. While preliminary analyses of the brain sections have been reported, a comprehensive microscopic neuroanatomical analysis of the state of H.M.'s brain at the time of his death has not yet been published. The brain tissue and slides were subsequently moved to the University of California Davis and the slides digitized at high resolution. Initial stages of producing a website for the public viewing of the images were also carried out. Recently, the slides, digital images, and tissue have been transferred to Boston University for permanent archiving. A new steering committee has been established and plans are in place for completion of a freely accessible H.M. website. Research publications on the microscopic anatomy and neuropathology of H.M.'s brain at the time of his death are also planned. We write this commentary to provide the hippocampus and memory neuroscience communities with a brief summary of what has transpired following H.M.'s death and outline plans for future publications and a tissue archive.


Asunto(s)
Encéfalo , Hipocampo , Humanos , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Imagen por Resonancia Magnética
2.
Cereb Cortex ; 32(10): 2170-2196, 2022 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-34613380

RESUMEN

The laminar cellular and circuit mechanisms by which the anterior cingulate cortex (ACC) exerts flexible control of motor and affective information for goal-directed behavior have not been elucidated. Using multimodal tract-tracing, in vitro patch-clamp recording and computational approaches in rhesus monkeys (M. mulatta), we provide evidence that specialized motor and affective network dynamics can be conferred by layer-specific biophysical and structural properties of ACC pyramidal neurons targeting two key downstream structures -the dorsal premotor cortex (PMd) and the amygdala (AMY). AMY-targeting neurons exhibited significant laminar differences, with L5 more excitable (higher input resistance and action potential firing rates) than L3 neurons. Between-pathway differences were found within L5, with AMY-targeting neurons exhibiting greater excitability, apical dendritic complexity, spine densities, and diversity of inhibitory inputs than PMd-targeting neurons. Simulations using a pyramidal-interneuron network model predict that these layer- and pathway-specific single-cell differences contribute to distinct network oscillatory dynamics. L5 AMY-targeting networks are more tuned to slow oscillations well-suited for affective and contextual processing timescales, while PMd-targeting networks showed strong beta/gamma synchrony implicated in rapid sensorimotor processing. These findings are fundamental to our broad understanding of how layer-specific cellular and circuit properties can drive diverse laminar activity found in flexible behavior.


Asunto(s)
Giro del Cíngulo , Corteza Prefrontal , Potenciales de Acción/fisiología , Dendritas , Giro del Cíngulo/fisiología , Corteza Prefrontal/fisiología , Células Piramidales/fisiología
3.
Mol Psychiatry ; 26(6): 1808-1831, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32071385

RESUMEN

Maternal immune activation (MIA) disrupts the central innate immune system during a critical neurodevelopmental period. Microglia are primary innate immune cells in the brain although their direct influence on the MIA phenotype is largely unknown. Here we show that MIA alters microglial gene expression with upregulation of cellular protrusion/neuritogenic pathways, concurrently causing repetitive behavior, social deficits, and synaptic dysfunction to layer V intrinsically bursting pyramidal neurons in the prefrontal cortex of mice. MIA increases plastic dendritic spines of the intrinsically bursting neurons and their interaction with hyper-ramified microglia. Treating MIA offspring by colony stimulating factor 1 receptor inhibitors induces depletion and repopulation of microglia, and corrects protein expression of the newly identified MIA-associated neuritogenic molecules in microglia, which coalesces with correction of MIA-associated synaptic, neurophysiological, and behavioral abnormalities. Our study demonstrates that maternal immune insults perturb microglial phenotypes and influence neuronal functions throughout adulthood, and reveals a potent effect of colony stimulating factor 1 receptor inhibitors on the correction of MIA-associated microglial, synaptic, and neurobehavioral dysfunctions.


Asunto(s)
Microglía , Efectos Tardíos de la Exposición Prenatal , Animales , Conducta Animal , Encéfalo , Modelos Animales de Enfermedad , Femenino , Inflamación , Factor Estimulante de Colonias de Macrófagos , Ratones , Neuronas , Embarazo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos
4.
Brain ; 144(1): 288-309, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33246331

RESUMEN

Extracellular vesicles are highly transmissible and play critical roles in the propagation of tau pathology, although the underlying mechanism remains elusive. Here, for the first time, we comprehensively characterized the physicochemical structure and pathogenic function of human brain-derived extracellular vesicles isolated from Alzheimer's disease, prodromal Alzheimer's disease, and non-demented control cases. Alzheimer's disease extracellular vesicles were significantly enriched in epitope-specific tau oligomers in comparison to prodromal Alzheimer's disease or control extracellular vesicles as determined by dot blot and atomic force microscopy. Alzheimer's disease extracellular vesicles were more efficiently internalized by murine cortical neurons, as well as more efficient in transferring and misfolding tau, than prodromal Alzheimer's disease and control extracellular vesicles in vitro. Strikingly, the inoculation of Alzheimer's disease or prodromal Alzheimer's disease extracellular vesicles containing only 300 pg of tau into the outer molecular layer of the dentate gyrus of 18-month-old C57BL/6 mice resulted in the accumulation of abnormally phosphorylated tau throughout the hippocampus by 4.5 months, whereas inoculation of an equal amount of tau from control extracellular vesicles, isolated tau oligomers, or fibrils from the same Alzheimer's disease donor showed little tau pathology. Furthermore, Alzheimer's disease extracellular vesicles induced misfolding of endogenous tau in both oligomeric and sarkosyl-insoluble forms in the hippocampal region. Unexpectedly, phosphorylated tau was primarily accumulated in glutamic acid decarboxylase 67 (GAD67) GABAergic interneurons and, to a lesser extent, glutamate receptor 2/3-positive excitatory mossy cells, showing preferential extracellular vesicle-mediated GABAergic interneuronal tau propagation. Whole-cell patch clamp recordings of CA1 pyramidal cells showed significant reduction in the amplitude of spontaneous inhibitory post-synaptic currents. This was accompanied by reductions in c-fos+ GAD67+ neurons and GAD67+ neuronal puncta surrounding pyramidal neurons in the CA1 region, confirming reduced GABAergic transmission in this region. Our study posits a novel mechanism for the spread of tau in hippocampal GABAergic interneurons via brain-derived extracellular vesicles and their subsequent neuronal dysfunction.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Vesículas Extracelulares/metabolismo , Interneuronas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Vesículas Extracelulares/patología , Femenino , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/patología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Interneuronas/patología , Masculino , Ratones Endogámicos C57BL , Células Piramidales/metabolismo , Células Piramidales/patología
5.
Am J Primatol ; 83(11): e23299, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34255875

RESUMEN

While humans exhibit a significant degree of neuropathological changes associated with deficits in cognitive and memory functions during aging, non-human primates (NHP) present with more variable expressions of pathological alterations among individuals and species. As such, NHP with long life expectancy in captivity offer an opportunity to study brain senescence in the absence of the typical cellular pathology caused by age-related neurodegenerative illnesses commonly seen in humans. Age-related changes at neuronal population, single cell, and synaptic levels have been well documented in macaques and marmosets, while age-related and Alzheimer's disease-like neuropathology has been characterized in additional species including lemurs as well as great apes. We present a comparative overview of existing neuropathologic observations across the primate order, including classic age-related changes such as cell loss, amyloid deposition, amyloid angiopathy, and tau accumulation. We also review existing cellular and ultrastructural data on neuronal changes, such as dendritic attrition and spine alterations, synaptic loss and pathology, and axonal and myelin pathology, and discuss their repercussions on cellular and systems function and cognition.


Asunto(s)
Envejecimiento , Encéfalo/patología , Primates , Enfermedad de Alzheimer , Animales , Angiopatía Amiloide Cerebral
6.
Cereb Cortex ; 29(3): 1121-1138, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29415216

RESUMEN

How the variety of neurons that organize into neocortical layers and functional areas arises is a central question in the study of cortical development. While both intrinsic and extrinsic cues are known to influence this process, whether distinct neuronal progenitor groups contribute to neuron diversity and allocation is poorly understood. Using in vivo genetic fate-mapping combined with whole-cell patch clamp recording, we show that the firing pattern and apical dendritic morphology of excitatory neurons in layer 4 of the barrel cortex are specified in part by their neural precursor lineage. Further, we show that separate precursors contribute to unique features of barrel cortex topography including the intralaminar position and thalamic innervation of the neurons they generate. Importantly, many of these lineage-specified characteristics are different from those previously measured for pyramidal neurons in layers 2-3 of the frontal cortex. Collectively, our data elucidate a dynamic temporal program in neuronal precursors that fine-tunes the properties of their progeny according to the lamina of destination.


Asunto(s)
Células-Madre Neurales/fisiología , Células Piramidales/fisiología , Corteza Somatosensorial/crecimiento & desarrollo , Potenciales de Acción , Animales , Espinas Dendríticas , Femenino , Masculino , Ratones , Modelos Neurológicos , Neocórtex/citología , Neocórtex/crecimiento & desarrollo , Células Piramidales/citología , Corteza Somatosensorial/citología , Proteínas de Dominio T Box/metabolismo
7.
J Neurosci ; 37(18): 4717-4734, 2017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28381592

RESUMEN

The lateral prefrontal cortex (LPFC) and anterior cingulate cortex (ACC) of the primate play distinctive roles in the mediation of complex cognitive tasks. Compared with the LPFC, integration of information by the ACC can span longer timescales and requires stronger engagement of inhibitory processes. Here, we reveal the synaptic mechanism likely to underlie these differences using in vitro patch-clamp recordings of synaptic events and multiscale imaging of synaptic markers in rhesus monkeys. Although excitatory synaptic signaling does not differ, the level of synaptic inhibition is much higher in ACC than LPFC layer 3 pyramidal neurons, with a significantly higher frequency (∼6×) and longer duration of inhibitory synaptic currents. The number of inhibitory synapses and the ratio of cholecystokinin to parvalbumin-positive inhibitory inputs are also significantly higher in ACC compared with LPFC neurons. Therefore, inhibition is functionally and structurally more robust and diverse in ACC than in LPFC, resulting in a lower excitatory: inhibitory ratio and a greater dynamic range for signal integration and network oscillation by the ACC. These differences in inhibitory circuitry likely underlie the distinctive network dynamics in ACC and LPC during normal and pathological brain states.SIGNIFICANCE STATEMENT The lateral prefrontal cortex (LPFC) and anterior cingulate cortex (ACC) play temporally distinct roles during the execution of cognitive tasks (rapid working memory during ongoing tasks and long-term memory to guide future action, respectively). Compared with LPFC-mediated tasks, ACC-mediated tasks can span longer timescales and require stronger engagement of inhibition. This study shows that inhibitory signaling is much more robust and diverse in the ACC than in the LPFC. Therefore, there is a lower excitatory: inhibitory synaptic ratio and a greater dynamic range for signal integration and oscillatory behavior in the ACC. These significant differences in inhibitory synaptic transmission form an important basis for the differential timing of cognitive processing by the LPFC and ACC in normal and pathological brain states.


Asunto(s)
Giro del Cíngulo/fisiología , Red Nerviosa/fisiología , Inhibición Neural/fisiología , Plasticidad Neuronal/fisiología , Corteza Prefrontal/fisiología , Transmisión Sináptica/fisiología , Animales , Femenino , Giro del Cíngulo/citología , Macaca mulatta , Masculino , Red Nerviosa/citología , Corteza Prefrontal/citología
8.
Cereb Cortex ; 27(3): 2078-2094, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26965903

RESUMEN

A principal challenge of systems neuroscience is to understand the unique characteristics of cortical neurons and circuits that enable area- and species-specific sensory encoding, motor function, cognition, and behavior. To address this issue, we compared properties of layer 3 pyramidal neurons in 2 cortical areas that span a broad range of cortical function-primary sensory (V1), to cognitive (frontal)-in the mouse and the rhesus monkey. Hierarchical clustering and discriminant analyses of 15 physiological and 25 morphological variables revealed 2 fundamental principles. First, V1 and frontal neurons are remarkably similar with regard to nearly every property in the mouse, while the opposite is true in the monkey, with V1 and frontal neurons exhibiting significant differences in nearly every property assessed. Second, neurons within visual and frontal areas differ significantly between the mouse and the monkey. Neurons in mouse and monkey V1 are the same size, but differ in nearly every other way; mouse frontal cortical neurons are smaller than those in the monkey and also differ substantially with regard to most other properties. These findings have broad implications for understanding the differential contributions of heterogeneous neuronal types in construction of cortical microcircuitry in diverse brain areas and species.


Asunto(s)
Lóbulo Frontal/citología , Macaca mulatta/anatomía & histología , Ratones/anatomía & histología , Células Piramidales/citología , Corteza Somatosensorial/citología , Animales , Tamaño de la Célula , Análisis por Conglomerados , Lóbulo Frontal/fisiología , Procesamiento de Imagen Asistido por Computador , Macaca mulatta/fisiología , Potenciales de la Membrana/fisiología , Ratones/fisiología , Microscopía Confocal , Técnicas de Placa-Clamp , Células Piramidales/fisiología , Corteza Somatosensorial/fisiología
9.
J Neurosci ; 35(1): 112-27, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25568107

RESUMEN

Understanding commonalities and differences in glutamatergic synaptic signaling is essential for understanding cortical functional diversity, especially in the highly complex primate brain. Previously, we have shown that spontaneous EPSCs differed markedly in layer 3 pyramidal neurons of two specialized cortical areas in the rhesus monkey, the high-order lateral prefrontal cortex (LPFC) and the primary visual cortex (V1). Here, we used patch-clamp recordings and confocal and electron microscopy to determine whether these distinct synaptic responses are due to differences in firing rates of presynaptic neurons and/or in the features of presynaptic or postsynaptic entities. As with spontaneous EPSCs, TTX-insensitive (action potential-independent) miniature EPSCs exhibited significantly higher frequency, greater amplitude, and slower kinetics in LPFC compared with V1 neurons. Consistent with these physiological differences, LPFC neurons possessed higher densities of spines, and the mean width of large spines was greater compared with those on V1 neurons. Axospinous synapses in layers 2-3 of LPFC had larger postsynaptic density surface areas and a higher proportion of large perforated synapses compared with V1. Axonal boutons in LPFC were also larger in volume and contained ∼ 1.6× more vesicles than did those in V1. Further, LPFC had a higher density of AMPA GluR2 receptor labeling than V1. The properties of spines and synaptic currents of individual layer 3 pyramidal neurons measured here were significantly correlated, consistent with the idea that significantly more frequent and larger synaptic currents are likely due to more numerous, larger, and more powerful synapses in LPFC compared with V1.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/fisiología , Corteza Prefrontal/fisiología , Sinapsis/fisiología , Corteza Visual/fisiología , Animales , Femenino , Macaca mulatta , Masculino , Técnicas de Cultivo de Órganos
10.
J Neurosci ; 35(15): 6142-52, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25878286

RESUMEN

Several neural precursor populations contemporaneously generate neurons in the developing neocortex. Specifically, radial glial stem cells of the dorsal telencephalon divide asymmetrically to produce excitatory neurons, but also indirectly to produce neurons via three types of intermediate progenitor cells. Why so many precursor types are needed to produce neurons has not been established; whether different intermediate progenitor cells merely expand the output of radial glia or instead generate distinct types of neurons is unknown. Here we use a novel genetic fate mapping technique to simultaneously track multiple precursor streams in the developing mouse brain and show that layer 2 and 3 pyramidal neurons exhibit distinctive electrophysiological and structural properties depending upon their precursor cell type of origin. These data indicate that individual precursor subclasses synchronously produce functionally different neurons, even within the same lamina, and identify a primary mechanism leading to cortical neuronal diversity.


Asunto(s)
Linaje de la Célula/fisiología , Neocórtex/citología , Red Nerviosa/fisiología , Células-Madre Neurales/clasificación , Células-Madre Neurales/fisiología , Células Piramidales/fisiología , Animales , Electroporación , Embrión de Mamíferos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Proteínas Luminiscentes/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Potenciales de la Membrana/fisiología , Ratones , Ratones Transgénicos , Neocórtex/embriología , Técnicas de Placa-Clamp , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
11.
J Comput Neurosci ; 41(1): 65-90, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27106692

RESUMEN

Conductance-based compartment modeling requires tuning of many parameters to fit the neuron model to target electrophysiological data. Automated parameter optimization via evolutionary algorithms (EAs) is a common approach to accomplish this task, using error functions to quantify differences between model and target. We present a three-stage EA optimization protocol for tuning ion channel conductances and kinetics in a generic neuron model with minimal manual intervention. We use the technique of Latin hypercube sampling in a new way, to choose weights for error functions automatically so that each function influences the parameter search to a similar degree. This protocol requires no specialized physiological data collection and is applicable to commonly-collected current clamp data and either single- or multi-objective optimization. We applied the protocol to two representative pyramidal neurons from layer 3 of the prefrontal cortex of rhesus monkeys, in which action potential firing rates are significantly higher in aged compared to young animals. Using an idealized dendritic topology and models with either 4 or 8 ion channels (10 or 23 free parameters respectively), we produced populations of parameter combinations fitting the target datasets in less than 80 hours of optimization each. Passive parameter differences between young and aged models were consistent with our prior results using simpler models and hand tuning. We analyzed parameter values among fits to a single neuron to facilitate refinement of the underlying model, and across fits to multiple neurons to show how our protocol will lead to predictions of parameter differences with aging in these neurons.


Asunto(s)
Envejecimiento/fisiología , Evolución Biológica , Canales Iónicos/fisiología , Modelos Neurológicos , Células Piramidales/fisiología , Potenciales de Acción , Algoritmos , Animales , Simulación por Computador , Dendritas , Técnicas In Vitro , Cinética , Macaca mulatta , Células Piramidales/citología
12.
Cereb Cortex ; 25(6): 1454-68, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24323499

RESUMEN

The effects of normal aging on morphologic and electrophysiologic properties of layer 3 pyramidal neurons in rhesus monkey primary visual cortex (V1) were assessed with whole-cell, patch-clamp recordings in in vitro slices. In another cohort of monkeys, the ultrastructure of synapses in the layers 2-3 neuropil of V1 was assessed using electron microscopy. Distal apical dendritic branching complexity was reduced in aged neurons, as was the total spine density, due to specific loss of mushroom spines from the apical tree and of thin spines from the basal tree. There was also an age-related decrease in the numerical density of symmetric and asymmetric synapses. In contrast to these structural changes, intrinsic membrane, action potential (AP), and excitatory and inhibitory synaptic current properties were the same in aged and young neurons. Computational modeling using morphologic reconstructions predicts that reduced dendritic complexity leads to lower attenuation of voltage outward from the soma (e.g., backpropagating APs) in aged neurons. Importantly, none of the variables that changed with age differed in neurons from cognitively impaired versus unimpaired aged monkeys. In summary, there are age-related alterations to the structural properties of V1 neurons, but these are not associated with significant electrophysiologic changes or with cognitive decline.


Asunto(s)
Envejecimiento , Cognición/fisiología , Células Piramidales/fisiología , Corteza Visual/citología , Animales , Simulación por Computador , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Lisina/análogos & derivados , Macaca mulatta , Masculino , Potenciales de la Membrana/fisiología , Microscopía Electrónica , Modelos Neurológicos , Pruebas Neuropsicológicas , Técnicas de Placa-Clamp , Células Piramidales/ultraestructura , Sinapsis/ultraestructura
13.
J Comput Neurosci ; 38(2): 263-83, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25527184

RESUMEN

Layer 3 (L3) pyramidal neurons in the lateral prefrontal cortex (LPFC) of rhesus monkeys exhibit dendritic regression, spine loss and increased action potential (AP) firing rates during normal aging. The relationship between these structural and functional alterations, if any, is unknown. To address this issue, morphological and electrophysiological properties of L3 LPFC pyramidal neurons from young and aged rhesus monkeys were characterized using in vitro whole-cell patch-clamp recordings and high-resolution digital reconstruction of neurons. Consistent with our previous studies, aged neurons exhibited significantly reduced dendritic arbor length and spine density, as well as increased input resistance and firing rates. Computational models using the digital reconstructions with Hodgkin-Huxley and AMPA channels allowed us to assess relationships between demonstrated age-related changes and to predict physiological changes that have not yet been tested empirically. For example, the models predict that in both backpropagating APs and excitatory postsynaptic currents (EPSCs), attenuation is lower in aged versus young neurons. Importantly, when identical densities of passive parameters and voltage- and calcium-gated conductances were used in young and aged model neurons, neither input resistance nor firing rates differed between the two age groups. Tuning passive parameters for each model predicted significantly higher membrane resistance (R m ) in aged versus young neurons. This R m increase alone did not account for increased firing rates in aged models, but coupling these R m values with subtle differences in morphology and membrane capacitance did. The predicted differences in passive parameters (or parameters with similar effects) are mathematically plausible, but must be tested empirically.


Asunto(s)
Potenciales de Acción/fisiología , Envejecimiento/fisiología , Dendritas , Potenciales Postsinápticos Excitadores/fisiología , Modelos Neurológicos , Células Piramidales/fisiología , Animales , Electrofisiología/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Macaca mulatta , Técnicas de Placa-Clamp/métodos , Corteza Prefrontal/citología , Receptores AMPA/fisiología , Canales de Sodio Activados por Voltaje/fisiología
14.
J Neurosci ; 33(6): 2408-18, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23392670

RESUMEN

Growing evidence suggests that a physiological activity of the cellular prion protein (PrP(C)) plays a crucial role in several neurodegenerative disorders, including prion and Alzheimer's diseases. However, how the functional activity of PrP(C) is subverted to deliver neurotoxic signals remains uncertain. Transgenic (Tg) mice expressing PrP with a deletion of residues 105-125 in the central region (referred to as ΔCR PrP) provide important insights into this problem. Tg(ΔCR) mice exhibit neonatal lethality and massive degeneration of cerebellar granule neurons, a phenotype that is dose dependently suppressed by the presence of wild-type PrP. When expressed in cultured cells, ΔCR PrP induces large, ionic currents that can be detected by patch-clamping techniques. Here, we tested the hypothesis that abnormal ion channel activity underlies the neuronal death seen in Tg(ΔCR) mice. We find that ΔCR PrP induces abnormal ionic currents in neurons in culture and in cerebellar slices and that this activity sensitizes the neurons to glutamate-induced, calcium-mediated death. In combination with ultrastructural and biochemical analyses, these results demonstrate a role for glutamate-induced excitotoxicity in PrP-mediated neurodegeneration. A similar mechanism may operate in other neurodegenerative disorders attributable to toxic, ß-rich oligomers that bind to PrP(C).


Asunto(s)
Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Glutámico/toxicidad , Ratones Endogámicos C57BL/fisiología , Mutación/fisiología , Neuronas/fisiología , Proteínas PrPC/biosíntesis , Animales , Células Cultivadas , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Proteínas PrPC/genética
15.
J Neurosci ; 33(5): 1927-39, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23365232

RESUMEN

We have previously shown that myelin abnormalities characterize the normal aging process of the brain and that an age-associated reduction in Klotho is conserved across species. Predominantly generated in brain and kidney, Klotho overexpression extends life span, whereas loss of Klotho accelerates the development of aging-like phenotypes. Although the function of Klotho in brain is unknown, loss of Klotho expression leads to cognitive deficits. We found significant effects of Klotho on oligodendrocyte functions, including induced maturation of rat primary oligodendrocytic progenitor cells (OPCs) in vitro and myelination. Phosphoprotein analysis indicated that Klotho's downstream effects involve Akt and ERK signal pathways. Klotho increased OPC maturation, and inhibition of Akt or ERK function blocked this effect on OPCs. In vivo studies of Klotho knock-out mice and control littermates revealed that knock-out mice have a significant reduction in major myelin protein and gene expression. By immunohistochemistry, the number of total and mature oligodendrocytes was significantly lower in Klotho knock-out mice. Strikingly, at the ultrastructural level, Klotho knock-out mice exhibited significantly impaired myelination of the optic nerve and corpus callosum. These mice also displayed severe abnormalities at the nodes of Ranvier. To decipher the mechanisms by which Klotho affects oligodendrocytes, we used luciferase pathway reporters to identify the transcription factors involved. Together, these studies provide novel evidence for Klotho as a key player in myelin biology, which may thus be a useful therapeutic target in efforts to protect brain myelin against age-dependent changes and promote repair in multiple sclerosis.


Asunto(s)
Encéfalo/metabolismo , Glucuronidasa/metabolismo , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Oligodendroglía/metabolismo , Animales , Recuento de Células , Supervivencia Celular/fisiología , Células Cultivadas , Cuerpo Calloso/metabolismo , Femenino , Glucuronidasa/genética , Proteínas Klotho , Ratones , Ratones Noqueados , Proteína Básica de Mielina/metabolismo , Células-Madre Neurales/metabolismo , Nervio Óptico/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT1/fisiología
16.
Brain Struct Funct ; 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38943018

RESUMEN

In this novel large-scale multiplexed immunofluorescence study we comprehensively characterized and compared layer-specific proteomic features within regions of interest of the widely divergent dorsolateral prefrontal cortex (A46) and primary visual cortex (A17) of adult rhesus monkeys. Twenty-eight markers were imaged in rounds of sequential staining, and their spatial distribution precisely quantified within gray matter layers and superficial white matter. Cells were classified as neurons, astrocytes, oligodendrocytes, microglia, or endothelial cells. The distribution of fibers and blood vessels were assessed by quantification of staining intensity across regions of interest. This method revealed multivariate similarities and differences between layers and areas. Protein expression in neurons was the strongest determinant of both laminar and regional differences, whereas protein expression in glia was more important for intra-areal laminar distinctions. Among specific results, we observed a lower glia-to-neuron ratio in A17 than in A46 and the pan-neuronal markers HuD and NeuN were differentially distributed in both brain areas with a lower intensity of NeuN in layers 4 and 5 of A17 compared to A46 and other A17 layers. Astrocytes and oligodendrocytes exhibited distinct marker-specific laminar distributions that differed between regions; notably, there was a high proportion of ALDH1L1-expressing astrocytes and of oligodendrocyte markers in layer 4 of A17. The many nuanced differences in protein expression between layers and regions observed here highlight the need for direct assessment of proteins, in addition to RNA expression, and set the stage for future protein-focused studies of these and other brain regions in normal and pathological conditions.

17.
J Neurosci ; 32(40): 13644-60, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23035077

RESUMEN

Whole-cell patch-clamp recordings and high-resolution 3D morphometric analyses of layer 3 pyramidal neurons in in vitro slices of monkey primary visual cortex (V1) and dorsolateral granular prefrontal cortex (dlPFC) revealed that neurons in these two brain areas possess highly distinctive structural and functional properties. Area V1 pyramidal neurons are much smaller than dlPFC neurons, with significantly less extensive dendritic arbors and far fewer dendritic spines. Relative to dlPFC neurons, V1 neurons have a significantly higher input resistance, depolarized resting membrane potential, and higher action potential (AP) firing rates. Most V1 neurons exhibit both phasic and regular-spiking tonic AP firing patterns, while dlPFC neurons exhibit only tonic firing. Spontaneous postsynaptic currents are lower in amplitude and have faster kinetics in V1 than in dlPFC neurons, but are no different in frequency. Three-dimensional reconstructions of V1 and dlPFC neurons were incorporated into computational models containing Hodgkin-Huxley and AMPA receptor and GABA(A) receptor gated channels. Morphology alone largely accounted for observed passive physiological properties, but led to AP firing rates that differed more than observed empirically, and to synaptic responses that opposed empirical results. Accordingly, modeling predicts that active channel conductances differ between V1 and dlPFC neurons. The unique features of V1 and dlPFC neurons are likely fundamental determinants of area-specific network behavior. The compact electrotonic arbor and increased excitability of V1 neurons support the rapid signal integration required for early processing of visual information. The greater connectivity and dendritic complexity of dlPFC neurons likely support higher level cognitive functions including working memory and planning.


Asunto(s)
Neuronas/fisiología , Corteza Prefrontal/fisiología , Células Piramidales/fisiología , Corteza Visual/fisiología , Potenciales de Acción , Animales , Espinas Dendríticas/fisiología , Espinas Dendríticas/ultraestructura , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/fisiología , Macaca mulatta , Masculino , Microscopía Confocal , Modelos Neurológicos , Neuronas/ultraestructura , Especificidad de Órganos , Técnicas de Placa-Clamp , Corteza Prefrontal/citología , Transmisión Sináptica , Corteza Visual/citología
18.
bioRxiv ; 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37693412

RESUMEN

Normal aging leads to myelin alternations in the rhesus monkey dorsolateral prefrontal cortex (dlPFC), which are often correlated with cognitive impairment. It is hypothesized that remyelination with shorter and thinner myelin sheaths partially compensates for myelin degradation, but computational modeling has not yet explored these two phenomena together systematically. Here, we used a two-pronged modeling approach to determine how age-related myelin changes affect a core cognitive function: spatial working memory. First we built a multicompartment pyramidal neuron model fit to monkey dlPFC data, with axon including myelinated segments having paranodes, juxtaparanodes, internodes, and tight junctions, to quantify conduction velocity (CV) changes and action potential (AP) failures after demyelination and subsequent remyelination in a population of neurons. Lasso regression identified distinctive parameter sets likely to modulate an axon's susceptibility to CV changes following demyelination versus remyelination. Next we incorporated the single neuron results into a spiking neural network model of working memory. While complete remyelination nearly recovered axonal transmission and network function to unperturbed levels, our models predict that biologically plausible levels of myelin dystrophy, if uncompensated by other factors, can account for substantial working memory impairment with aging. The present computational study unites empirical data from electron microscopy up to behavior on aging, and has broader implications for many demyelinating conditions, such as multiple sclerosis or schizophrenia.

19.
bioRxiv ; 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36909556

RESUMEN

Much is known about differences in pyramidal cells across cortical areas and species, but studies of interneurons have focused on comparisons within single cortical areas and/or species. Here we quantified the distribution and somato-dendritic morphology of interneurons expressing one or more of the calcium binding proteins (CaBPs) calretinin (CR), calbindin (CB) and/or parvalbumin (PV) in mouse ( Mus musculus ) versus rhesus monkey ( Macaca mulatta ) in two functionally and cytoarchitectonically distinct regions- the primary visual and frontal cortical areas. The density, laminar distribution and morphology of interneurons were assessed in serial brain sections using immunofluorescent multi-labeling, stereological counting and 3D reconstructions. There were significantly higher densities of CB+ and PV+ neurons in visual compared to frontal areas in both species. The main species difference was the significantly greater density and proportion of CR+ interneurons and lower extent of CaBP co-expression in monkey compared to mouse cortices. Cluster analyses revealed that the somato-dendritic morphology of layer 2-3 inhibitory interneurons is more dependent on CaBP expression than on species and area. Only modest effects of species were observed for CB+ and PV+ interneuron morphologies, while CR+ neurons showed no difference. By contrast to pyramidal cells which show highly distinctive area- and species-specific features, here we found more subtle differences in the distribution and features of interneurons across areas and species. These data yield insight into how nuanced differences in the population organization and properties of neurons may underlie specializations in cortical regions to confer species and area-specific functional capacities. Key Points: Somato-dendritic morphology of distinct interneurons did not substantially scale and vary across areas and species- differences were mainly dependent on CaBP expression.Cortical diversity in inhibitory function across areas and species is thus likely to be derived from differential laminar distribution and densities of distinct interneuron subclasses.In contrast to pyramidal cells which differ widely in distribution and morphology across areas and species, the features of interneurons appears to be relatively more conserved across areas and species.

20.
J Comp Neurol ; 531(18): 1934-1962, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37357562

RESUMEN

Fundamental differences in excitatory pyramidal cells across cortical areas and species highlight the implausibility of extrapolation from mouse to primate neurons and cortical networks. Far less is known about comparative regional and species-specific features of neurochemically distinct cortical inhibitory interneurons. Here, we quantified the density, laminar distribution, and somatodendritic morphology of inhibitory interneurons expressing one or more of the calcium-binding proteins (CaBPs) (calretinin [CR], calbindin [CB], and/or parvalbumin [PV]) in mouse (Mus musculus) versus rhesus monkey (Macaca mulatta) in two functionally and cytoarchitectonically distinct regions-the primary visual and frontal cortical areas-using immunofluorescent multilabeling, stereological counting, and 3D reconstructions. There were significantly higher densities of CB+ and PV+ neurons in visual compared to frontal areas in both species. The main species difference was the significantly greater density and proportion of CR+ interneurons and lower extent of CaBP coexpression in monkey compared to mouse cortices. Cluster analyses revealed that the somatodendritic morphology of layer 2-3 inhibitory interneurons is more dependent on CaBP expression than on species and area. Only modest effects of species were observed for CB+ and PV+ interneuron morphologies, while CR+ neurons showed no difference. By contrast to pyramidal cells that show highly distinctive area- and species-specific features, here we found more subtle differences in the distribution and features of interneurons across areas and species. These data yield insight into how nuanced differences in the population organization and properties of neurons may underlie specializations in cortical regions to confer species- and area-specific functional capacities.


Asunto(s)
Parvalbúminas , Proteína G de Unión al Calcio S100 , Animales , Ratones , Calbindinas/metabolismo , Calbindina 2/metabolismo , Parvalbúminas/metabolismo , Proteína G de Unión al Calcio S100/análisis , Proteína G de Unión al Calcio S100/metabolismo , Corteza Prefrontal , Interneuronas/metabolismo , Lóbulo Frontal , Macaca mulatta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA