Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 7(1): 605, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769398

RESUMEN

Alzheimer's disease (AD) is broadly characterized by neurodegeneration, pathology accumulation, and cognitive decline. There is considerable variation in the progression of clinical symptoms and pathology in humans, highlighting the importance of genetic diversity in the study of AD. To address this, we analyze cell composition and amyloid-beta deposition of 6- and 14-month-old AD-BXD mouse brains. We utilize the analytical QUINT workflow- a suite of software designed to support atlas-based quantification, which we expand to deliver a highly effective method for registering and quantifying cell and pathology changes in diverse disease models. In applying the expanded QUINT workflow, we quantify near-global age-related increases in microglia, astrocytes, and amyloid-beta, and we identify strain-specific regional variation in neuron load. To understand how individual differences in cell composition affect the interpretation of bulk gene expression in AD, we combine hippocampal immunohistochemistry analyses with bulk RNA-sequencing data. This approach allows us to categorize genes whose expression changes in response to AD in a cell and/or pathology load-dependent manner. Ultimately, our study demonstrates the use of the QUINT workflow to standardize the quantification of immunohistochemistry data in diverse mice, - providing valuable insights into regional variation in cellular load and amyloid deposition in the AD-BXD model.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Modelos Animales de Enfermedad , Variación Genética , Animales , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Ratones , Encéfalo/metabolismo , Encéfalo/patología , Ratones Transgénicos , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/genética , Masculino
2.
Sci Adv ; 10(14): eadj9305, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38569042

RESUMEN

The power and scope of disease modeling can be markedly enhanced through the incorporation of broad genetic diversity. The introduction of pathogenic mutations into a single inbred mouse strain sometimes fails to mimic human disease. We describe a cross-species precision disease modeling platform that exploits mouse genetic diversity to bridge cell-based modeling with whole organism analysis. We developed a universal protocol that permitted robust and reproducible neural differentiation of genetically diverse human and mouse pluripotent stem cell lines and then carried out a proof-of-concept study of the neurodevelopmental gene DYRK1A. Results in vitro reliably predicted the effects of genetic background on Dyrk1a loss-of-function phenotypes in vivo. Transcriptomic comparison of responsive and unresponsive strains identified molecular pathways conferring sensitivity or resilience to Dyrk1a1A loss and highlighted differential messenger RNA isoform usage as an important determinant of response. This cross-species strategy provides a powerful tool in the functional analysis of candidate disease variants identified through human genetic studies.


Asunto(s)
Células Madre Pluripotentes , Animales , Ratones , Humanos , Fenotipo
3.
Alzheimers Dement (N Y) ; 10(1): e12458, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38469553

RESUMEN

INTRODUCTION: In September 2022, The Jackson Laboratory Center for Alzheimer's and Dementia Research (JAX CADR) hosted a workshop with leading researchers in the Alzheimer's disease and related dementias (ADRD) field. METHODS: During the workshop, the participants brainstormed new directions to overcome current barriers to providing patients with effective ADRD therapeutics. The participants outlined specific areas of focus. Following the workshop, each group used standard literature search methods to provide background for each topic. RESULTS: The team of invited experts identified four key areas that can be collectively addressed to make a significant impact in the field: (1) Prioritize the diversification of disease targets, (2) enhance factors promoting resilience, (3) de-risk clinical pipeline, and (4) centralize data management. DISCUSSION: In this report, we review these four objectives and propose innovations to expedite ADRD therapeutic pipelines.

4.
Int J Obes (Lond) ; 47(3): 224-235, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36725979

RESUMEN

BACKGROUND/OBJECTIVE: As the obesity epidemic continues, the understanding of macronutrient influence on central nervous system function is critical for understanding diet-induced obesity and potential therapeutics, particularly in light of the increased sugar content in processed foods. Previous research showed mixed effects of sucrose feeding on body weight gain but has yet to reveal insight into the impact of sucrose on hypothalamic functioning. Here, we explore the impact of liquid sucrose feeding for 12 weeks on body weight, body composition, caloric intake, and hypothalamic AgRP neuronal function and synaptic plasticity. METHODS: Patch-clamp electrophysiology of hypothalamic AgRP neurons, metabolic phenotyping and food intake were performed on C57BL/6J mice. RESULTS: While mice given sugar-sweetened water do not gain significant weight, they do show subtle differences in body composition and caloric intake. When given sugar-sweetened water, mice show similar alterations to AgRP neuronal excitability as in high-fat diet obese models. Increased sugar consumption also primes mice for increased caloric intake and weight gain when given access to a HFD. CONCLUSIONS: Our results show that elevated sucrose consumption increased activity of AgRP neurons and altered synaptic excitability. This may contribute to obesity in mice and humans with access to more palatable (HFD) diets.


Asunto(s)
Obesidad , Sacarosa , Humanos , Ratones , Animales , Sacarosa/farmacología , Sacarosa/metabolismo , Proteína Relacionada con Agouti/metabolismo , Ratones Endogámicos C57BL , Aumento de Peso , Dieta Alta en Grasa , Neuronas/metabolismo , Agua/metabolismo , Agua/farmacología , Peso Corporal
5.
Trends Neurosci ; 45(5): 369-383, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35307206

RESUMEN

Individuals who maintain cognitive function despite high levels of Alzheimer's disease (AD)-associated pathology are said to be 'resilient' to AD. Identifying mechanisms underlying resilience represents an exciting therapeutic opportunity. Human studies have identified a number of molecular and genetic factors associated with resilience, but the complexity of these cohorts prohibits a complete understanding of which factors are causal or simply correlated with resilience. Genetically and phenotypically diverse mouse models of AD provide new and translationally relevant opportunities to identify and prioritize new resilience mechanisms for further cross-species investigation. This review will discuss insights into resilience gained from both human and animal studies and highlight future approaches that may help translate these insights into therapeutics designed to prevent or delay AD-related dementia.


Asunto(s)
Enfermedad de Alzheimer , Resiliencia Psicológica , Animales , Encéfalo , Cognición , Humanos , Ratones , Procesamiento Proteico-Postraduccional
6.
Front Cell Dev Biol ; 8: 562662, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042997

RESUMEN

Developing strategies to maintain cognitive health is critical to quality of life during aging. The basis of healthy cognitive aging is poorly understood; thus, it is difficult to predict who will have normal cognition later in life. Individuals may have higher baseline functioning (cognitive reserve) and others may maintain or even improve with age (cognitive resilience). Understanding the mechanisms underlying cognitive reserve and resilience may hold the key to new therapeutic strategies for maintaining cognitive health. However, reserve and resilience have been inconsistently defined in human studies. Additionally, our understanding of the molecular and cellular bases of these phenomena is poor, compounded by a lack of longitudinal molecular and cognitive data that fully capture the dynamic trajectories of cognitive aging. Here, we used a genetically diverse mouse population (B6-BXDs) to characterize individual differences in cognitive abilities in adulthood and investigate evidence of cognitive reserve and/or resilience in middle-aged mice. We tested cognitive function at two ages (6 months and 14 months) using y-maze and contextual fear conditioning. We observed heritable variation in performance on these traits (h 2 RIx̄ = 0.51-0.74), suggesting moderate to strong genetic control depending on the cognitive domain. Due to the polygenetic nature of cognitive function, we did not find QTLs significantly associated with y-maze, contextual fear acquisition (CFA) or memory, or decline in cognitive function at the genome-wide level. To more precisely interrogate the molecular regulation of variation in these traits, we employed RNA-seq and identified gene networks related to transcription/translation, cellular metabolism, and neuronal function that were associated with working memory, contextual fear memory, and cognitive decline. Using this method, we nominate the Trio gene as a modulator of working memory ability. Finally, we propose a conceptual framework for identifying strains exhibiting cognitive reserve and/or resilience to assess whether these traits can be observed in middle-aged B6-BXDs. Though we found that earlier cognitive reserve evident early in life protects against cognitive impairment later in life, cognitive performance and age-related decline fell along a continuum, with no clear genotypes emerging as exemplars of exceptional reserve or resilience - leading to recommendations for future use of aging mouse populations to understand the nature of cognitive reserve and resilience.

7.
Learn Mem ; 27(9): 355-371, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32817302

RESUMEN

Individual differences in cognitive decline during normal aging and Alzheimer's disease (AD) are common, but the molecular mechanisms underlying these distinct outcomes are not fully understood. We utilized a combination of genetic, molecular, and behavioral data from a mouse population designed to model human variation in cognitive outcomes to search for the molecular mechanisms behind this population-wide variation. Specifically, we used a systems genetics approach to relate gene expression to cognitive outcomes during AD and normal aging. Statistical causal-inference Bayesian modeling was used to model systematic genetic perturbations matched with cognitive data that identified astrocyte and microglia molecular networks as drivers of cognitive resilience to AD. Using genetic mapping, we identified Fgf2 as a potential regulator of the astrocyte network associated with individual differences in short-term memory. We also identified several immune genes as regulators of a microglia network associated with individual differences in long-term memory, which was partly mediated by amyloid burden. Finally, significant overlap between mouse and two different human coexpression networks provided strong evidence of translational relevance for the genetically diverse AD-BXD panel as a model of late-onset AD. Together, this work identified two candidate molecular pathways enriched for microglia and astrocyte genes that serve as causal AD cognitive biomarkers, and provided a greater understanding of processes that modulate individual and population-wide differences in cognitive outcomes during AD.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer , Astrocitos , Disfunción Cognitiva , Reserva Cognitiva , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Microglía , Envejecimiento/genética , Envejecimiento/inmunología , Envejecimiento/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Conducta Animal/fisiología , Biomarcadores , Encéfalo , Disfunción Cognitiva/genética , Disfunción Cognitiva/inmunología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Reserva Cognitiva/fisiología , Femenino , Humanos , Individualidad , Masculino , Ratones , Ratones Transgénicos , Modelos Genéticos
8.
Genes Brain Behav ; 18(8): e12603, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31381246

RESUMEN

Many patients with Alzheimer's dementia (AD) also exhibit noncognitive symptoms such as sensorimotor deficits, which can precede the hallmark cognitive deficits and significantly impact daily activities and an individual's ability to live independently. However, the mechanisms underlying sensorimotor dysfunction in AD and their relationship with cognitive decline remains poorly understood, due in part to a lack of translationally relevant animal models. To address this, we recently developed a novel model of genetic diversity in Alzheimer's disease, the AD-BXD genetic reference panel. In this study, we investigated sensorimotor deficits in the AD-BXDs and the relationship to cognitive decline in these mice. We found that age- and AD-related declines in coordination, balance and vestibular function vary significantly across the panel, indicating genetic background strongly influences the expressivity of the familial AD mutations used in the AD-BXD panel and their impact on motor function. Although young males and females perform comparably regardless of genotype on narrow beam and inclined screen tasks, there were significant sex differences in aging- and AD-related decline, with females exhibiting worse decline than males of the same age and transgene status. Finally, we found that AD motor decline is not correlated with cognitive decline, suggesting that sensorimotor deficits in AD may occur through distinct mechanisms. Overall, our results suggest that AD-related sensorimotor decline is strongly dependent on background genetics and is independent of dementia and cognitive deficits, suggesting that effective therapeutics for the entire spectrum of AD symptoms will likely require interventions targeting each distinct domain involved in the disease.


Asunto(s)
Enfermedad de Alzheimer/genética , Genotipo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Animales , Cognición , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Destreza Motora , Movimiento , Factores Sexuales
9.
Neurosci Biobehav Rev ; 103: 73-80, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31207254

RESUMEN

Diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) arise from complex interactions of genetic and environmental factors, with genetic variants regulating individual responses to environmental exposures (i.e. gene-by-environment interactions). Identifying gene-by-environment interactions will be critical to fully understanding disease mechanisms and developing personalized therapeutics, though these interactions are still poorly understood and largely under-studied. Candidate gene approaches have shown that known disease risk variants often regulate response to environmental factors. However, recent improvements in exposome- and genome-wide association and interaction studies in humans and mice are enabling discovery of novel genetic variants and pathways that predict response to a variety of environmental factors. Here, we highlight recent approaches and ongoing developments in human and rodent studies to identify genetic modulators of environmental factors using AD and PD as exemplars. Identifying gene-by-environment interactions in disease will be critical to developing personalized intervention strategies and will pave the way for precision medicine.


Asunto(s)
Enfermedad de Alzheimer , Exposoma , Interacción Gen-Ambiente , Estudio de Asociación del Genoma Completo , Enfermedad de Parkinson , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/genética , Animales , Humanos , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética
10.
Neuron ; 101(3): 399-411.e5, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30595332

RESUMEN

An individual's genetic makeup plays a large role in determining susceptibility to Alzheimer's disease (AD) but has largely been ignored in preclinical studies. To test the hypothesis that incorporating genetic diversity into mouse models of AD would improve translational potential, we combined a well-established mouse model of AD with a genetically diverse reference panel to generate mice that harbor identical high-risk human mutations but differ across the remainder of their genome. We first show that genetic variation profoundly modifies the impact of human AD mutations on both cognitive and pathological phenotypes. We then validate this complex AD model by demonstrating high degrees of genetic, transcriptomic, and phenotypic overlap with human AD. Overall, work here both introduces a novel AD mouse population as an innovative and reproducible resource for the study of mechanisms underlying AD and provides evidence that preclinical models incorporating genetic diversity may better translate to human disease.


Asunto(s)
Enfermedad de Alzheimer/genética , Modelos Animales de Enfermedad , Medicina de Precisión/métodos , Investigación Biomédica Traslacional/normas , Enfermedad de Alzheimer/patología , Animales , Femenino , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Medicina de Precisión/normas , Especificidad de la Especie , Investigación Biomédica Traslacional/métodos
11.
eNeuro ; 5(6)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30627661

RESUMEN

The subiculum is the main target of the hippocampal region CA1 and is the principle output region of the hippocampus. The subiculum is critical to learning and memory, although it has been relatively understudied. There are two functional types of principle neurons within the subiculum: regular spiking (RS) and burst spiking (BS) neurons. To determine whether these cell types are differentially modified by learning-related experience, we performed whole-cell patch clamp recordings from male mouse brain slices following contextual fear conditioning (FC) and memory retrieval relative to a number of control behavioral paradigms. RS cells, but not BS cells, displayed a greater degree of experience-related plasticity in intrinsic excitability measures [afterhyperpolarization (AHP), input resistance (Rinput), current required to elicit a spike], with fear conditioned animals having generally more excitable RS cells compared to naïve controls. Furthermore, we found that the relative proportion of RS to BS neurons is modified by the type of exposure, with the lowest proportion of BS subicular cells occurring in animals that underwent contextual FC followed by a retrieval test. These studies indicate that pyramidal neurons in the subiculum undergo experience- and learning-related plasticity in intrinsic properties in a cell-type-specific manner. As BS and RS cells are thought to convey distinct types of information, this plasticity may be particularly important in encoding, consolidating, and recalling spatial information by modulating information flow from the hippocampus to cortical regions.


Asunto(s)
Condicionamiento Clásico/fisiología , Ambiente , Miedo/fisiología , Hipocampo/citología , Neuronas/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Condicionamiento Clásico/efectos de los fármacos , Estimulación Eléctrica , Electrochoque , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Hipocampo/fisiología , Técnicas In Vitro , Ácido Quinurénico/farmacología , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/clasificación , Neuronas/efectos de los fármacos , Piridazinas/farmacología
12.
J Tissue Eng Regen Med ; 12(4): e1836-e1851, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29139614

RESUMEN

Induced pluripotent stem cells (iPSCs) give rise to neural stem/progenitor cells, serving as a good source for neural regeneration. Here, we established transgene-free (TF) iPSCs from dental stem cells (DSCs) and determined their capacity to differentiate into functional neurons in vitro. Generated TF iPSCs from stem cells of apical papilla and dental pulp stem cells underwent two methods-embryoid body-mediated and direct induction, to guide TF-DSC iPSCs along with H9 or H9 Syn-GFP (human embryonic stem cells) into functional neurons in vitro. Using the embryoid body-mediated method, early stage neural markers PAX6, SOX1, and nestin were detected by immunocytofluorescence or reverse transcription-real time polymerase chain reaction (RT-qPCR). At late stage of neural induction measured at Weeks 7 and 9, the expression levels of neuron-specific markers Nav1.6, Kv1.4, Kv4.2, synapsin, SNAP25, PSD95, GAD67, GAP43, and NSE varied between stem cells of apical papilla iPSCs and H9. For direct induction method, iPSCs were directly induced into neural stem/progenitor cells and guided to become neuron-like cells. The direct method, while simpler, showed cell detachment and death during the differentiation process. At early stage, PAX6, SOX1 and nestin were detected. At late stage of differentiation, all five genes tested, nestin, ßIII-tubulin, neurofilament medium chain, GFAP, and Nav, were positive in many cells in cultures. Both differentiation methods led to neuron-like cells in cultures exhibiting sodium and potassium currents, action potential, or spontaneous excitatory postsynaptic potential. Thus, TF-DSC iPSCs are capable of undergoing guided neurogenic differentiation into functional neurons in vitro, thereby may serve as a cell source for neural regeneration.


Asunto(s)
Diferenciación Celular , Pulpa Dental/metabolismo , Cuerpos Embrioides/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Adolescente , Adulto , Antígenos de Diferenciación/biosíntesis , Pulpa Dental/citología , Cuerpos Embrioides/citología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Células-Madre Neurales/citología , Neuronas/citología
13.
Neurobiol Aging ; 46: 58-67, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27460150

RESUMEN

An individual's genetic makeup plays an important role in determining susceptibility to cognitive aging. Identifying the specific genes that contribute to cognitive aging may aid in early diagnosis of at-risk patients, as well as identify novel therapeutics targets to treat or prevent development of symptoms. Challenges to identifying these specific genes in human studies include complex genetics, difficulty in controlling environmental factors, and limited access to human brain tissue. Here, we identify Hp1bp3 as a novel modulator of cognitive aging using a genetically diverse population of mice and confirm that HP1BP3 protein levels are significantly reduced in the hippocampi of cognitively impaired elderly humans relative to cognitively intact controls. Deletion of functional Hp1bp3 in mice recapitulates memory deficits characteristic of aged impaired mice and humans, further supporting the idea that Hp1bp3 and associated molecular networks are modulators of cognitive aging. Overall, our results suggest Hp1bp3 may serve as a potential target against cognitive aging and demonstrate the utility of genetically diverse animal models for the study of complex human disease.


Asunto(s)
Envejecimiento/genética , Trastornos del Conocimiento/genética , Cognición/fisiología , Envejecimiento Cognitivo/fisiología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad/genética , Trastornos de la Memoria/genética , Memoria/fisiología , Proteínas Nucleares/fisiología , Animales , Trastornos del Conocimiento/psicología , Condicionamiento Psicológico/fisiología , Modelos Animales de Enfermedad , Miedo , Femenino , Humanos , Masculino , Trastornos de la Memoria/psicología , Ratones , Ratones Noqueados
14.
Sci Rep ; 5: 16810, 2015 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-26592769

RESUMEN

Obesity is a chronic condition resulting from a long-term pattern of poor diet and lifestyle. Long-term consumption of high-fat diet (HFD) leads to persistent activation and leptin resistance in AgRP neurons in the arcuate nucleus of the hypothalamus (ARH). Here, for the first time, we demonstrate acute effects of HFD on AgRP neuronal excitability and highlight a critical role for diet composition. In parallel with our earlier finding in obese, long-term HFD mice, we found that even brief HFD feeding results in persistent activation of ARH AgRP neurons. However, unlike long-term HFD-fed mice, AgRP neurons from short-term HFD-fed mice were still leptin-sensitive, indicating that the development of leptin-insensitivity is not a prerequisite for the increased firing rate of AgRP neurons. To distinguish between diet composition, caloric intake, and body weight, we compared acute and long-term effects of HFD and CD in pair-fed mice on AgRP neuronal spiking. HFD consumption in pair-fed mice resulted in a significant increase in AgRP neuronal spiking despite controls for weight gain and caloric intake. Taken together, our results suggest that diet composition may be more important than either calorie intake or body weight for electrically remodeling arcuate AgRP/NPY neurons.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Proteína Relacionada con Agouti/genética , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Grasas de la Dieta/efectos adversos , Neuropéptido Y/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/fisiopatología , Dieta Alta en Grasa/efectos adversos , Ingestión de Energía , Regulación de la Expresión Génica , Leptina/genética , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Transducción de Señal
15.
J Neurosci ; 34(16): 5486-96, 2014 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-24741039

RESUMEN

The hypothalamic arcuate nucleus (ARH) is a brain region critical for regulation of food intake and a primary area for the action of leptin in the CNS. In lean mice, the adipokine leptin inhibits neuropeptide Y (NPY) and agouti-related peptide (AgRP) neuronal activity, resulting in decreased food intake. Here we show that diet-induced obesity in mice is associated with persistent activation of NPY neurons and a failure of leptin to reduce the firing rate or hyperpolarize the resting membrane potential. However, the molecular mechanism whereby diet uncouples leptin's effect on neuronal excitability remains to be fully elucidated. In NPY neurons from lean mice, the Kv channel blocker 4-aminopyridine inhibited leptin-induced changes in input resistance and spike rate. Consistent with this, we found that ARH NPY neurons have a large, leptin-sensitive delayed rectifier K(+) current and that leptin sensitivity of this current is blunted in neurons from diet-induced obese mice. This current is primarily carried by Kv2-containing channels, as the Kv2 channel inhibitor stromatoxin-1 significantly increased the spontaneous firing rate in NPY neurons from lean mice. In HEK cells, leptin induced a significant hyperpolarizing shift in the voltage dependence of Kv2.1 but had no effect on the function of the closely related channel Kv2.2 when these channels were coexpressed with the long isoform of the leptin receptor LepRb. Our results suggest that dynamic modulation of somatic Kv2.1 channels regulates the intrinsic excitability of NPY neurons to modulate the spontaneous activity and the integration of synaptic input onto these neurons in the ARH.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Núcleo Arqueado del Hipotálamo/citología , Leptina/farmacología , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Obesidad/inducido químicamente , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Proteína Relacionada con Agouti/genética , Animales , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Neuropéptido Y/genética , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Shab/metabolismo
16.
Proc Natl Acad Sci U S A ; 107(27): 12351-6, 2010 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-20566856

RESUMEN

The Kv2.1 K(+) channel is highly expressed throughout the brain, where it regulates excitability during periods of high-frequency stimulation. Kv2.1 is unique among Kv channels in that it targets to large surface clusters on the neuronal soma and proximal dendrites. These clusters also form in transfected HEK cells. Following excessive excitatory stimulation, Kv2.1 declusters with an accompanying 20- to 30-mV hyperpolarizing shift in the activation threshold. Although most Kv2.1 channels are clustered, there is a pool of Kv2.1 resident outside of these domains. Using the cell-attached patch clamp technique, we investigated the hypothesis that Kv2.1 activity varies as a function of cell surface location. We found that clustered Kv2.1 channels do not efficiently conduct K(+), whereas the nonclustered channels are responsible for the high threshold delayed rectifier K(+) current typical of Kv2.1. Comparison of gating and ionic currents indicates only 2% of the surface channels conduct, suggesting that the clustered channels still respond to membrane potential changes. Declustering induced via either actin depolymerization or alkaline phosphatase treatment did not increase whole-cell currents. Dephosphorylation resulted in a 25-mV hyperpolarizing shift, whereas actin depolymerization did not alter the activation midpoint. Taken together, these data demonstrate that clusters do not contain high threshold Kv2.1 channels whose voltage sensitivity shifts upon declustering; nor are they a reservoir of nonconducting channels that are activated upon release. On the basis of these findings, we propose unique roles for the clustered Kv2.1 that are independent of K(+) conductance.


Asunto(s)
Activación del Canal Iónico/fisiología , Potasio/metabolismo , Canales de Potasio Shab/fisiología , Algoritmos , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Potenciales de la Membrana , Técnicas de Placa-Clamp , Puntos Cuánticos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo , Transfección
17.
Am J Physiol Heart Circ Physiol ; 294(1): H229-37, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17965280

RESUMEN

The delayed-rectifier voltage-gated K(+) channel (Kv) 2.1 underlies the cardiac slow K(+) current in the rodent heart and is particularly interesting in that both its function and localization are regulated by many stimuli in neuronal systems. However, standard immunolocalization approaches do not detect cardiac Kv2.1; therefore, little is known regarding its localization in the heart. In the present study, we used recombinant adenovirus to determine the subcellular localization and lateral mobility of green fluorescent protein (GFP)-Kv2.1 and yellow fluorescent protein-Kv1.4 in atrial and ventricular myocytes. In atrial myocytes, Kv2.1 formed large clusters on the cell surface similar to those observed in hippocampal neurons, whereas Kv1.4 was evenly distributed over both the peripheral sarcolemma and the transverse tubules. However, fluorescence recovery after photobleach (FRAP) experiments indicate that atrial Kv2.1 was immobile, whereas Kv1.4 was mobile (tau = 252 +/- 42 s). In ventricular myocytes, Kv2.1 did not form clusters and was localized primarily in the transverse-axial tubules and sarcolemma. In contrast, Kv1.4 was found only in transverse tubules and sarcolemma. FRAP studies revealed that Kv2.1 has a higher mobility in ventricular myocytes (tau = 479 +/- 178 s), although its mobility is slower than Kv1.4 (tau(1) = 18.9 +/- 2.3 s; tau(2) = 305 +/- 55 s). We also observed the movement of small, intracellular transport vesicles containing GFP-Kv2.1 within ventricular myocytes. These data are the first evidence of Kv2.1 localization in living myocytes and indicate that Kv2.1 may have distinct physiological roles in atrial and ventricular myocytes.


Asunto(s)
Canal de Potasio Kv.1.2/metabolismo , Canal de Potasio Kv1.4/metabolismo , Miocitos Cardíacos/metabolismo , Adenoviridae/genética , Animales , Línea Celular , Células Cultivadas , Recuperación de Fluorescencia tras Fotoblanqueo , Vectores Genéticos , Proteínas Fluorescentes Verdes , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Humanos , Canal de Potasio Kv.1.2/genética , Canal de Potasio Kv1.4/genética , Masculino , Microscopía Confocal , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Sarcolema/metabolismo , Factores de Tiempo , Transfección , Vesículas Transportadoras/metabolismo
18.
J Cell Sci ; 120(Pt 14): 2413-23, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17606996

RESUMEN

The Kv2.1 delayed-rectifier channel trafficks to 1-3 microm(2) clusters on the surface of neurons and transfected HEK cells. Single quantum dot (Qdot) tracking and FRAP approaches were used to quantify the diffusion of GFP-labeled Kv2.1 channels on the cell surface and address the mechanisms underlying the formation of these unique membrane structures. Mean square displacement analysis of single Kv2.1 channel tracks inside or outside the surface clusters yielded mean diffusion coefficients of 0.03+/-0.02 microm(2)/second and 0.06+/-0.05 microm(2)/second, respectively. Kv2.1 channels outside the clusters effectively ignore the cluster boundary, readily diffusing through these microdomains. However, in 5% of the tracks analyzed, single, non-clustered channels were observed to cross into a cluster and become corralled within the cluster perimeter. Alexa Fluor 594-labelled phalloidin staining and mCherry-Kv2.1 co-expression with GFP-actin indicated that the Kv2.1 surface clusters form where the cortical actin cytoskeleton is reduced. Kv2.1 channels lacking the C-terminus do not form clusters, freely diffusing over the cell surface with a mean diffusion coefficient of 0.07+/-0.04 microm(2)/second. These data support a model whereby the Kv2.1 clusters are formed by sub-membrane cytoskeletal structures that limit the lateral diffusion of only the sub-population of Kv2.1 channels carrying the appropriate modifications on the Kv2.1 C-terminus.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Citoesqueleto/metabolismo , Canales de Potasio Shab/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular/fisiología , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Humanos , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología
19.
J Neurosci ; 26(38): 9609-18, 2006 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-16988031

RESUMEN

Ion channel localization to specific cell surface regions is essential for proper neuronal function. The Kv2.1 K+ channel forms large clusters on the plasma membrane of hippocampal neurons and transfected human embryonic kidney (HEK) cells. Using live cell imaging, we address mechanisms underlying this Kv2.1 clustering in both HEK cells and cultured hippocampal neurons. The Kv2.1-containing surface clusters have properties unlike those expected for a scaffolding protein bound channel. After channel is delivered to the plasma membrane via intracellular transport vesicles, it remains localized at the insertion site. Fluorescence recovery after photobleaching (FRAP) and quantum dot tracking experiments indicate that channel within the surface cluster is mobile (FRAP, tau = 14.1 +/- 1.5 and 11.5 +/- 6.1 s in HEK cells and neurons, respectively). The cluster perimeter is not static, because after fusion of adjacent clusters, green fluorescent protein (GFP)-Kv2.1 completely exchanged between the two domains within 60 s. Treatment of hippocampal neurons expressing GFP-Kv2.1 with 5 microM latrunculin A resulted in a significant increase in average cluster size from 0.89 +/- 0.16 microm2 to 12.15 +/- 1.4 microm2 with a concomitant decrease in cluster number. Additionally, Kv2.1 was no longer restricted to the cell body, suggesting a role for cortical actin in both cluster maintenance and localization. Thus, Kv2.1 surface domains likely trap mobile Kv2.1 channels within a well defined, but fluid, perimeter rather than being tightly bound to a scaffolding protein-containing complex. Channel moves directly into these clusters via trafficking vesicles. Such domains allow for efficient trafficking to the cell surface while sequestering channel with signaling proteins.


Asunto(s)
Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Canales de Potasio Shab/química , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Humanos , Datos de Secuencia Molecular , Neuronas/química , Neuronas/metabolismo , Transporte de Proteínas/fisiología , Ratas , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo
20.
J Cell Sci ; 118(Pt 10): 2155-66, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15855232

RESUMEN

Voltage-gated potassium (Kv) channels regulate action potential duration in nerve and muscle; therefore changes in the number and location of surface channels can profoundly influence electrical excitability. To investigate trafficking of Kv2.1, 1.4 and 1.3 within the plasma membrane, we combined the expression of fluorescent protein-tagged Kv channels with live cell confocal imaging. Kv2.1 exhibited a clustered distribution in HEK cells similar to that seen in hippocampal neurons, whereas Kv1.4 and Kv1.3 were evenly distributed over the plasma membrane. Using FRAP, surface Kv2.1 displayed limited mobility; approximately 40% of the fluorescence recovered within 20 minutes of photobleach (M(f)=0.41+/-0.04). Recovery occurred not by diffusion from adjacent membrane but probably by transport of nascent channel from within the cell. By contrast, the Kv1 family members Kv1.4 and Kv1.3 were highly mobile, both showing approximately 80% recovery (Kv 1.4 M(f)=0.78+/-0.07; Kv1.3 M(f)=0.78+/-0.04; without correction for photobleach); unlike Kv2.1, recovery was consistent with diffusion of channel from membrane adjacent to the bleach region. Studies using PA-GFP-tagged channels were consistent with the FRAP results. Following photoactivation of a small region of plasma membrane PA-GFP-Kv2.1 remained restricted to the photoactivation ROI, while PA-GFP-Kv1.4 rapidly diffused throughout the cell surface. Additionally, PA-GFP-Kv2.1 moved into regions of the cell membrane not adjacent to the original photoactivation ROI. Sucrose density gradient analysis indicated that half of Kv2.1 is part of a large, macromolecular complex while Kv1.4 sediments as predicted for the tetrameric channel complex. Disruption of membrane cholesterol by cyclodextrin minimally altered Kv2.1 mobility (M(f)=0.32+/-0.03), but significantly increased surface cluster size by at least fourfold. By comparison, the mobility of Kv1.4 decreased following cholesterol depletion with no change in surface distribution. The mobility of Kv1.3 was slightly increased following cyclodextrin treatment. These results indicate that (1) Kv2.1, Kv1.4 and Kv1.3 exist in distinct compartments that exhibit different trafficking properties, (2) membrane cholesterol levels differentially modulate the trafficking and localization of Kv channels and (3) Kv2.1 expressed in HEK cells exhibits a surface distribution similar to that seen in native cells.


Asunto(s)
Membrana Celular/metabolismo , Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv1.4/metabolismo , Canales de Potasio Shab/metabolismo , Línea Celular , Colesterol/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Microdominios de Membrana/metabolismo , Isoformas de Proteínas/metabolismo , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...