Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 43(10): 1797-1813, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36746627

RESUMO

Despite the indispensable role that astrocytes play in the neurovascular unit, few studies have investigated the functional impact of astrocyte signaling in cognitive decline and dementia related to vascular pathology. Diet-mediated induction of hyperhomocysteinemia (HHcy) recapitulates numerous features of vascular contributions to cognitive impairment and dementia (VCID). Here, we used astrocyte targeting approaches to evaluate astrocyte Ca2+ dysregulation and the impact of aberrant astrocyte signaling on cerebrovascular dysfunction and synapse impairment in male and female HHcy diet mice. Two-photon imaging conducted in fully awake mice revealed activity-dependent Ca2+ dysregulation in barrel cortex astrocytes under HHcy. Stimulation of contralateral whiskers elicited larger Ca2+ transients in individual astrocytes of HHcy diet mice compared with control diet mice. However, evoked Ca2+ signaling across astrocyte networks was impaired in HHcy mice. HHcy also was associated with increased activation of the Ca2+/calcineurin-dependent transcription factor NFAT4, which has been linked previously to the reactive astrocyte phenotype and synapse dysfunction in amyloid and brain injury models. Targeting the NFAT inhibitor VIVIT to astrocytes, using adeno-associated virus vectors, led to reduced GFAP promoter activity in HHcy diet mice and improved functional hyperemia in arterioles and capillaries. VIVIT expression in astrocytes also preserved CA1 synaptic function and improved spontaneous alternation performance on the Y maze. Together, the results demonstrate that aberrant astrocyte signaling can impair the major functional properties of the neurovascular unit (i.e., cerebral vessel regulation and synaptic regulation) and may therefore represent a promising drug target for treating VCID and possibly Alzheimer's disease and other related dementias.SIGNIFICANCE STATEMENT The impact of reactive astrocytes in Alzheimer's disease and related dementias is poorly understood. Here, we evaluated Ca2+ responses and signaling in barrel cortex astrocytes of mice fed with a B-vitamin deficient diet that induces hyperhomocysteinemia (HHcy), cerebral vessel disease, and cognitive decline. Multiphoton imaging in awake mice with HHcy revealed augmented Ca2+ responses in individual astrocytes, but impaired signaling across astrocyte networks. Stimulation-evoked arteriole dilation and elevated red blood cell velocity in capillaries were also impaired in cortex of awake HHcy mice. Astrocyte-specific inhibition of the Ca2+-dependent transcription factor, NFAT, normalized cerebrovascular function in HHcy mice, improved synaptic properties in brain slices, and stabilized cognition. Results suggest that astrocytes are a mechanism and possible therapeutic target for vascular-related dementia.


Assuntos
Doença de Alzheimer , Hiper-Homocisteinemia , Camundongos , Masculino , Feminino , Animais , Doença de Alzheimer/metabolismo , Astrócitos/metabolismo , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Dieta , Fatores de Transcrição/metabolismo
2.
J Alzheimers Dis ; 78(4): 1419-1438, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33164928

RESUMO

BACKGROUND: In animal models and tissue preparations, calcium dyshomeostasis is a biomarker of aging and Alzheimer's disease that is associated with synaptic dysfunction, neuritic pruning, and dysregulated cellular processes. It is unclear, however, whether the onset of calcium dysregulation precedes, is concurrent with, or is the product of pathological cellular events (e.g., oxidation, amyloid-ß production, and neuroinflammation). Further, neuronal calcium dysregulation is not always present in animal models of amyloidogenesis, questioning its reliability as a disease biomarker. OBJECTIVE: Here, we directly tested for the presence of calcium dysregulation in dorsal hippocampal neurons in male and female 5×FAD mice on a C57BL/6 genetic background using sharp electrodes coupled with Oregon-green Bapta-1 imaging. We focused on three ages that coincide with the course of amyloid deposition: 1.5, 4, and 10 months old. METHODS: Outcome variables included measures of the afterhyperpolarization, short-term synaptic plasticity, and calcium kinetics during synaptic activation. Quantitative analyses of spatial learning and memory were also conducted using the Morris water maze. Main effects of sex, age, and genotype were identified on measures of electrophysiology and calcium imaging. RESULTS: Measures of resting Oregon-green Bapta-1 fluorescence showed significant reductions in the 5×FAD group compared to controls. Deficits in spatial memory, along with increases in Aß load, were detectable at older ages, allowing us to test for temporal associations with the onset of calcium dysregulation. CONCLUSION: Our results provide evidence that reduced, rather than elevated, neuronal calcium is identified in this 5×FAD model and suggests that this surprising result may be a novel biomarker of AD.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Cálcio/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Feminino , Hipocampo/citologia , Hipocampo/fisiopatologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Teste do Labirinto Aquático de Morris , Plasticidade Neuronal , Imagem Óptica , Técnicas de Patch-Clamp , Placa Amiloide/fisiopatologia , Presenilina-1/genética , Fatores Sexuais , Aprendizagem Espacial , Memória Espacial
3.
J Alzheimers Dis ; 77(4): 1623-1637, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32925058

RESUMO

BACKGROUND: Dysregulated signaling in neurons and astrocytes participates in pathophysiological alterations seen in the Alzheimer's disease brain, including increases in amyloid-ß, hyperphosphorylated tau, inflammation, calcium dysregulation, and oxidative stress. These are often noted prior to the development of behavioral, cognitive, and non-cognitive deficits. However, the extent to which these pathological changes function together or independently is unclear. OBJECTIVE: Little is known about the temporal relationship between calcium dysregulation and oxidative stress, as some reports suggest that dysregulated calcium promotes increased formation of reactive oxygen species, while others support the opposite. Prior work has quantified several key outcome measures associated with oxidative stress in aldehyde dehydrogenase 2 knockout (Aldh2-/-) mice, a non-transgenic model of sporadic Alzheimer's disease. METHODS: Here, we tested the hypothesis that early oxidative stress can promote calcium dysregulation across aging by measuring calcium-dependent processes using electrophysiological and imaging methods and focusing on the afterhyperpolarization (AHP), synaptic activation, somatic calcium, and long-term potentiation in the Aldh2-/- mouse. RESULTS: Our results show a significant age-related decrease in the AHP along with an increase in the slow AHP amplitude in Aldh2-/- animals. Measures of synaptic excitability were unaltered, although significant reductions in long-term potentiation maintenance were noted in the Aldh2-/- animals compared to wild-type. CONCLUSION: With so few changes in calcium and calcium-dependent processes in an animal model that shows significant increases in HNE adducts, Aß, p-tau, and activated caspases across age, the current findings do not support a direct link between neuronal calcium dysregulation and uncontrolled oxidative stress.


Assuntos
Aldeído-Desidrogenase Mitocondrial/deficiência , Doença de Alzheimer/metabolismo , Cálcio/metabolismo , Modelos Animais de Doenças , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Fatores Etários , Aldeído-Desidrogenase Mitocondrial/genética , Doença de Alzheimer/genética , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Imagem Molecular/métodos , Neurônios/química , Técnicas de Cultura de Órgãos
4.
J Alzheimers Dis ; 66(4): 1371-1378, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30412490

RESUMO

Aging is the leading risk factor for idiopathic Alzheimer's disease (AD), indicating that normal aging processes promote AD and likely are present in the neurons in which AD pathogenesis originates. In AD, neurofibrillary tangles (NFTs) appear first in entorhinal cortex, implying that aging processes in entorhinal neurons promote NFT pathogenesis. Using electrophysiology and immunohistochemistry, we find pronounced aging-related Ca2 + dysregulation in rat entorhinal neurons homologous with the human neurons in which NFTs originate. Considering that humans recapitulate many aspects of animal brain aging, these results support the hypothesis that aging-related Ca2 + dysregulation occurs in human entorhinal neurons and promotes NFT pathogenesis.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Córtex Entorrinal/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/patologia , Animais , Córtex Entorrinal/patologia , Masculino , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Neurônios/patologia , Ratos , Ratos Endogâmicos F344
5.
J Neurosci ; 38(4): 1030-1041, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29255009

RESUMO

Hippocampal overexpression of FK506-binding protein 12.6/1b (FKBP1b), a negative regulator of ryanodine receptor Ca2+ release, reverses aging-induced memory impairment and neuronal Ca2+ dysregulation. Here, we tested the hypothesis that FKBP1b also can protect downstream transcriptional networks from aging-induced dysregulation. We gave hippocampal microinjections of FKBP1b-expressing viral vector to male rats at either 13 months of age (long-term, LT) or 19 months of age (short-term, ST) and tested memory performance in the Morris water maze at 21 months of age. Aged rats treated ST or LT with FKBP1b substantially outperformed age-matched vector controls and performed similarly to each other and young controls (YCs). Transcriptional profiling in the same animals identified 2342 genes with hippocampal expression that was upregulated/downregulated in aged controls (ACs) compared with YCs (the aging effect). Of these aging-dependent genes, 876 (37%) also showed altered expression in aged FKBP1b-treated rats compared with ACs, with FKBP1b restoring expression of essentially all such genes (872/876, 99.5%) in the direction opposite the aging effect and closer to levels in YCs. This inverse relationship between the aging and FKBP1b effects suggests that the aging effects arise from FKBP1b deficiency. Functional category analysis revealed that genes downregulated with aging and restored by FKBP1b were associated predominantly with diverse brain structure categories, including cytoskeleton, membrane channels, and extracellular region. Conversely, genes upregulated with aging but not restored by FKBP1b associated primarily with glial-neuroinflammatory, ribosomal, and lysosomal categories. Immunohistochemistry confirmed aging-induced rarefaction and FKBP1b-mediated restoration of neuronal microtubular structure. Therefore, a previously unrecognized genomic network modulating diverse brain structural processes is dysregulated by aging and restored by FKBP1b overexpression.SIGNIFICANCE STATEMENT Previously, we found that hippocampal overexpression of FK506-binding protein 12.6/1b (FKBP1b), a negative regulator of intracellular Ca2+ responses, reverses both aging-related Ca2+ dysregulation and cognitive impairment. Here, we tested whether hippocampal FKBP1b overexpression also counteracts aging changes in gene transcriptional networks. In addition to reducing memory deficits in aged rats, FKBP1b selectively counteracted aging-induced expression changes in 37% of aging-dependent genes, with cytoskeletal and extracellular structure categories highly associated with the FKBP1b-rescued genes. Our results indicate that, in parallel with cognitive processes, a novel transcriptional network coordinating brain structural organization is dysregulated with aging and restored by FKBP1b.


Assuntos
Envelhecimento/fisiologia , Regulação da Expressão Gênica/fisiologia , Hipocampo/metabolismo , Memória/fisiologia , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Hipocampo/fisiopatologia , Masculino , Transtornos da Memória/fisiopatologia , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos
6.
FASEB J ; 31(9): 4179-4186, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28592637

RESUMO

Hippocampus oxidative stress is considered pathogenic in neurodegenerative diseases, such as Alzheimer disease (AD), and in neurodevelopmental disorders, such as Angelman syndrome (AS). Yet clinical benefits of antioxidant treatment for these diseases remain unclear because conventional imaging methods are unable to guide management of therapies in specific hippocampus subfields in vivo that underlie abnormal behavior. Excessive production of paramagnetic free radicals in nonhippocampus brain tissue can be measured in vivo as a greater-than-normal 1/T1 that is quenchable with antioxidant as measured by quench-assisted (Quest) MRI. Here, we further test this approach in phantoms, and we present proof-of-concept data in models of AD-like and AS hippocampus oxidative stress that also exhibit impaired spatial learning and memory. AD-like models showed an abnormal gradient along the CA1 dorsal-ventral axis of excessive free radical production as measured by Quest MRI, and redox-sensitive calcium dysregulation as measured by manganese-enhanced MRI and electrophysiology. In the AS model, abnormally high free radical levels were observed in dorsal and ventral CA1. Quest MRI is a promising in vivo paradigm for bridging brain subfield oxidative stress and behavior in animal models and in human patients to better manage antioxidant therapy in devastating neurodegenerative and neurodevelopmental diseases.-Berkowitz, B. A., Lenning, J., Khetarpal, N., Tran, C., Wu, J. Y., Berri, A. M., Dernay, K., Haacke, E. M., Shafie-Khorassani, F., Podolsky, R. H., Gant, J. C., Maimaiti, S., Thibault, O., Murphy, G. G., Bennett, B. M., Roberts, R. In vivo imaging of prodromal hippocampus CA1 subfield oxidative stress in models of Alzheimer disease and Angelman syndrome.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Síndrome de Angelman/diagnóstico por imagem , Região CA1 Hipocampal/patologia , Estresse Oxidativo/fisiologia , Sintomas Prodrômicos , Aldeído-Desidrogenase Mitocondrial/genética , Aldeído-Desidrogenase Mitocondrial/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Síndrome de Angelman/patologia , Animais , Antioxidantes , Cálcio/metabolismo , Radicais Livres , Imageamento por Ressonância Magnética/métodos , Manganês , Memória/fisiologia , Camundongos Knockout , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
7.
Biochem Biophys Res Commun ; 483(4): 981-987, 2017 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-27553276

RESUMO

Neuroscientists studying normal brain aging, spinal cord injury, Alzheimer's disease (AD) and other neurodegenerative diseases have focused considerable effort on carefully characterizing intracellular perturbations in calcium dynamics or levels. At the cellular level, calcium is known for controlling life and death and orchestrating most events in between. For many years, intracellular calcium has been recognized as an essential ion associated with nearly all cellular functions from cell growth to degeneration. Often the emphasis is on the negative impact of calcium dysregulation and the typical worse-case-scenario leading inevitably to cell death. However, even high amplitude calcium transients, when executed acutely, can alter neuronal communication and synaptic strength in positive ways, without necessarily killing neurons. Here, we focus on the evidence that calcium has a subtle and distinctive role in shaping and controlling synaptic events that underpin neuronal communication and that these subtle changes in aging or AD may contribute to cognitive decline. We emphasize that calcium imaging in dendritic components is ultimately necessary to directly test for the presence of age- or disease-associated alterations during periods of synaptic activation.


Assuntos
Encéfalo/fisiologia , Cálcio/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Modelos Biológicos
8.
J Gerontol A Biol Sci Med Sci ; 71(1): 30-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25659889

RESUMO

Peripheral insulin resistance is a key component of metabolic syndrome associated with obesity, dyslipidemia, hypertension, and type 2 diabetes. While the impact of insulin resistance is well recognized in the periphery, it is also becoming apparent in the brain. Recent studies suggest that insulin resistance may be a factor in brain aging and Alzheimer's disease (AD) whereby intranasal insulin therapy, which delivers insulin to the brain, improves cognition and memory in AD patients. Here, we tested a clinically relevant delivery method to determine the impact of two forms of insulin, short-acting insulin lispro (Humalog) or long-acting insulin detemir (Levemir), on cognitive functions in aged F344 rats. We also explored insulin effects on the Ca(2+)-dependent hippocampal afterhyperpolarization (AHP), a well-characterized neurophysiological marker of aging which is increased in the aged, memory impaired animal. Low-dose intranasal insulin improved memory recall in aged animals such that their performance was similar to that seen in younger animals. Further, because ex vivo insulin also reduced the AHP, our results suggest that the AHP may be a novel cellular target of insulin in the brain, and improved cognitive performance following intranasal insulin therapy may be the result of insulin actions on the AHP.


Assuntos
Envelhecimento , Encéfalo , Senescência Celular/fisiologia , Cognição , Insulina Detemir , Insulina Lispro , Administração Intranasal , Envelhecimento/metabolismo , Envelhecimento/psicologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Senescência Celular/efeitos dos fármacos , Cognição/efeitos dos fármacos , Cognição/fisiologia , Transtornos Cognitivos/metabolismo , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/metabolismo , Insulina Detemir/administração & dosagem , Insulina Detemir/metabolismo , Insulina Lispro/administração & dosagem , Insulina Lispro/metabolismo , Resistência à Insulina , Memória/efeitos dos fármacos , Ratos , Resultado do Tratamento
9.
J Neurosci ; 35(30): 10878-87, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26224869

RESUMO

Brain Ca2+ regulatory processes are altered during aging, disrupting neuronal, and cognitive functions. In hippocampal pyramidal neurons, the Ca2+ -dependent slow afterhyperpolarization (sAHP) exhibits an increase with aging, which correlates with memory impairment. The increased sAHP results from elevated L-type Ca2+ channel activity and ryanodine receptor (RyR)-mediated Ca2+ release, but underlying molecular mechanisms are poorly understood. Previously, we found that expression of the gene encoding FK506-binding protein 12.6/1b (FKBP1b), a small immunophilin that stabilizes RyR-mediated Ca2+ release in cardiomyocytes, declines in hippocampus of aged rats and Alzheimer's disease subjects. Additionally, knockdown/disruption of hippocampal FKBP1b in young rats augments neuronal Ca2+ responses. Here, we test the hypothesis that declining FKBP1b underlies aging-related hippocampal Ca2+ dysregulation. Using microinjection of adeno-associated viral vector bearing a transgene encoding FKBP1b into the hippocampus of aged male rats, we assessed the critical prediction that overexpressing FKBP1b should reverse Ca2+ -mediated manifestations of brain aging. Immunohistochemistry and qRT-PCR confirmed hippocampal FKBP1b overexpression 4-6 weeks after injection. Compared to aged vector controls, aged rats overexpressing FKBP1b showed dramatic enhancement of spatial memory, which correlated with marked reduction of sAHP magnitude. Furthermore, simultaneous electrophysiological recording and Ca2+ imaging in hippocampal neurons revealed that the sAHP reduction was associated with a decrease in parallel RyR-mediated Ca2+ transients. Thus, hippocampal FKBP1b overexpression reversed key aspects of Ca2+ dysregulation and cognitive impairment in aging rats, supporting the novel hypothesis that declining FKBP1b is a molecular mechanism underlying aging-related Ca2+ dysregulation and unhealthy brain aging and pointing to FKBP1b as a potential therapeutic target. SIGNIFICANCE STATEMENT: This paper reports critical tests of a novel hypothesis that proposes a molecular mechanism of unhealthy brain aging and possibly, Alzheimer's disease. For more than 30 years, evidence has been accumulating that brain aging is associated with dysregulation of calcium in neurons. Recently, we found that FK506-binding protein 12.6/1b (FKBP1b), a small protein that regulates calcium, declines with aging in the hippocampus, a brain region important for memory. Here we used gene therapy approaches and found that raising FKBP1b reversed calcium dysregulation and memory impairment in aging rats, allowing them to perform a memory task as well as young rats. These studies identify a potential molecular mechanism of brain aging and may also have implications for treatment of Alzheimer's disease.


Assuntos
Envelhecimento/fisiologia , Cálcio/metabolismo , Cognição/fisiologia , Neurônios/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Transgenes
10.
J Neurosci ; 32(46): 16129-40, 2012 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-23152597

RESUMO

Astrocytes are the most abundant cell type in the brain and play a critical role in maintaining healthy nervous tissue. In Alzheimer's disease (AD) and most other neurodegenerative disorders, many astrocytes convert to a chronically "activated" phenotype characterized by morphologic and biochemical changes that appear to compromise protective properties and/or promote harmful neuroinflammatory processes. Activated astrocytes emerge early in the course of AD and become increasingly prominent as clinical and pathological symptoms progress, but few studies have tested the potential of astrocyte-targeted therapeutics in an intact animal model of AD. Here, we used adeno-associated virus (AAV) vectors containing the astrocyte-specific Gfa2 promoter to target hippocampal astrocytes in APP/PS1 mice. AAV-Gfa2 vectors drove the expression of VIVIT, a peptide that interferes with the immune/inflammatory calcineurin/NFAT (nuclear factor of activated T-cells) signaling pathway, shown by our laboratory and others to orchestrate biochemical cascades leading to astrocyte activation. After several months of treatment with Gfa2-VIVIT, APP/PS1 mice exhibited improved cognitive and synaptic function, reduced glial activation, and lower amyloid levels. The results confirm a deleterious role for activated astrocytes in AD and lay the groundwork for exploration of other novel astrocyte-based therapies.


Assuntos
Doença de Alzheimer/patologia , Astrócitos/fisiologia , Animais , Astrócitos/patologia , Astrócitos/ultraestrutura , Aprendizagem da Esquiva/fisiologia , Western Blotting , Encéfalo/patologia , Inibidores de Calcineurina , Células Cultivadas , Dependovirus/genética , Ensaio de Imunoadsorção Enzimática , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas de Transferência de Genes , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Inflamação/fisiopatologia , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Transgênicos , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/fisiologia , Neurônios/fisiologia , Oligopeptídeos/farmacologia , Transdução de Sinais/fisiologia
11.
J Neurosci ; 31(5): 1693-703, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21289178

RESUMO

With aging, multiple Ca(2+)-associated electrophysiological processes exhibit increased magnitude in hippocampal pyramidal neurons, including the Ca(2+)-dependent slow afterhyperpolarization (sAHP), L-type voltage-gated Ca(2+) channel (L-VGCC) activity, Ca(2+)-induced Ca(2+) release (CICR) from ryanodine receptors (RyRs), and Ca(2+) transients. This pattern of Ca(2+) dysregulation correlates with reduced neuronal excitability/plasticity and impaired learning/memory and has been proposed to contribute to unhealthy brain aging and Alzheimer's disease. However, little is known about the underlying molecular mechanisms. In cardiomyocytes, FK506-binding protein 1b/12.6 (FKBP1b) binds and stabilizes RyR2 in the closed state, inhibiting RyR-mediated Ca(2+) release. Moreover, we recently found that hippocampal Fkbp1b expression is downregulated, whereas Ryr2 and Frap1/Mtor (mammalian target of rapamycin) expression is upregulated with aging in rats. Here, we tested the hypothesis that disrupting FKBP1b function also destabilizes Ca(2+) homeostasis in hippocampal neurons and is sufficient to induce the aging phenotype of Ca(2+) dysregulation in young animals. Selective knockdown of Fkbp1b with interfering RNA in vitro (96 h) enhanced voltage-gated Ca(2+) current in cultured neurons, whereas in vivo Fkbp1b knockdown by microinjection of viral vector (3-4 weeks) dramatically increased the sAHP in hippocampal slice neurons from young-adult rats. Rapamycin, which displaces FKBP1b from RyRs in myocytes, similarly enhanced VGCC current and the sAHP and also increased CICR. Moreover, FKBP1b knockdown in vivo was associated with upregulation of RyR2 and mTOR protein expression. Thus, disruption of FKBP1b recapitulated much of the Ca(2+)-dysregulation aging phenotype in young rat hippocampus, supporting a novel hypothesis that declining FKBP function plays a major role in unhealthy brain aging.


Assuntos
Envelhecimento/metabolismo , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio , Cálcio/metabolismo , Hipocampo/fisiopatologia , Células Piramidais/fisiopatologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Envelhecimento/genética , Animais , Células Cultivadas , Eletrofisiologia , Técnicas de Silenciamento de Genes , Vetores Genéticos , Hipocampo/metabolismo , Homeostase/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Potenciais da Membrana/efeitos dos fármacos , Microinjeções , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Células Piramidais/metabolismo , Ratos , Ratos Endogâmicos F344 , Sirolimo/farmacologia , Proteínas de Ligação a Tacrolimo/genética
12.
PLoS One ; 5(4): e10405, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20454453

RESUMO

BACKGROUND: Thiazolidinediones (TZDs) activate peroxisome proliferator-activated receptor gamma (PPARgamma) and are used clinically to help restore peripheral insulin sensitivity in Type 2 diabetes (T2DM). Interestingly, long-term treatment of mouse models of Alzheimer's disease (AD) with TZDs also has been shown to reduce several well-established brain biomarkers of AD including inflammation, oxidative stress and Abeta accumulation. While TZD's actions in AD models help to elucidate the mechanisms underlying their potentially beneficial effects in AD patients, little is known about the functional consequences of TZDs in animal models of normal aging. Because aging is a common risk factor for both AD and T2DM, we investigated whether the TZD, pioglitazone could alter brain aging under non-pathological conditions. METHODS AND FINDINGS: We used the F344 rat model of aging, and monitored behavioral, electrophysiological, and molecular variables to assess the effects of pioglitazone (PIO-Actos(R) a TZD) on several peripheral (blood and liver) and central (hippocampal) biomarkers of aging. Starting at 3 months or 17 months of age, male rats were treated for 4-5 months with either a control or a PIO-containing diet (final dose approximately 2.3 mg/kg body weight/day). A significant reduction in the Ca(2+)-dependent afterhyperpolarization was seen in the aged animals, with no significant change in long-term potentiation maintenance or learning and memory performance. Blood insulin levels were unchanged with age, but significantly reduced by PIO. Finally, a combination of microarray analyses on hippocampal tissue and serum-based multiplex cytokine assays revealed that age-dependent inflammatory increases were not reversed by PIO. CONCLUSIONS: While current research efforts continue to identify the underlying processes responsible for the progressive decline in cognitive function seen during normal aging, available medical treatments are still very limited. Because TZDs have been shown to have benefits in age-related conditions such as T2DM and AD, our study was aimed at elucidating PIO's potentially beneficial actions in normal aging. Using a clinically-relevant dose and delivery method, long-term PIO treatment was able to blunt several indices of aging but apparently affected neither age-related cognitive decline nor peripheral/central age-related increases in inflammatory signaling.


Assuntos
Envelhecimento/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Doença de Alzheimer , Animais , Biomarcadores/análise , Biomarcadores/sangue , Encéfalo/fisiologia , Hipocampo , Hipoglicemiantes/farmacologia , Inflamação , Aprendizagem/efeitos dos fármacos , Fígado , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Pioglitazona , Ratos
13.
J Neurosci ; 29(6): 1805-16, 2009 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-19211887

RESUMO

Multiple hippocampal processes and cognitive functions change with aging or Alzheimer's disease, but the potential triggers of these aging cascades are not well understood. Here, we quantified hippocampal expression profiles and behavior across the adult lifespan to identify early aging changes and changes that coincide with subsequent onset of cognitive impairment. Well powered microarray analyses (N = 49 arrays), immunohistochemistry, and Morris spatial maze learning were used to study male F344 rats at five age points. Genes that changed with aging (by ANOVA) were assigned to one of four onset age ranges based on template pattern matching; functional pathways represented by these genes were identified statistically (Gene Ontology). In the earliest onset age range (3-6 months old), upregulation began for genes in lipid/protein catabolic and lysosomal pathways, indicating a shift in metabolic substrates, whereas downregulation began for lipid synthesis, GTP/ATP-dependent signaling, and neural development genes. By 6-9 months of age, upregulation of immune/inflammatory cytokines was pronounced. Cognitive impairment first appeared in the midlife range (9-12 months) and coincided and correlated primarily with midlife upregulation of genes associated with cholesterol trafficking (apolipoprotein E), myelinogenic, and proteolytic/major histocompatibility complex antigen-presenting pathways. Immunolabeling revealed that cholesterol trafficking proteins were substantially increased in astrocytes and that myelination increased with aging. Together, our data suggest a novel sequential model in which an early-adult metabolic shift, favoring lipid/ketone body oxidation, triggers inflammatory degradation of myelin and resultant excess cholesterol that, by midlife, activates cholesterol transport from astrocytes to remyelinating oligodendrocytes. These processes may damage structure and compete with neuronal pathways for bioenergetic resources, thereby impairing cognitive function.


Assuntos
Envelhecimento/metabolismo , Colesterol/metabolismo , Cognição/fisiologia , Hipocampo/metabolismo , Transtornos da Memória/metabolismo , Animais , Animais Recém-Nascidos , Transtornos Cognitivos/metabolismo , Metabolismo Energético/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/etiologia , Ratos , Ratos Endogâmicos F344
14.
Epilepsia ; 50(4): 629-45, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18657176

RESUMO

PURPOSE: Clinically, perturbations in the semaphorin signaling system have been associated with autism and epilepsy. The semaphorins have been implicated in guidance, migration, differentiation, and synaptic plasticity of neurons. The semaphorin 3F (Sema3F) ligand and its receptor, neuropilin 2 (NPN2) are highly expressed within limbic areas. NPN2 signaling may intimately direct the apposition of presynaptic and postsynaptic locations, facilitating the development and maturity of hippocampal synaptic function. To further understand the role of NPN2 signaling in central nevous system (CNS) plasticity, structural and functional alterations were assessed in NPN2 deficient mice. METHODS: In NPN2 deficient mice, we measured seizure susceptibility after kainic acid or pentylenetetrazol, neuronal excitability and synaptic throughput in slice preparations, principal and interneuron cell counts with immunocytochemical protocols, synaptosomal protein levels with immunoblots, and dendritic morphology with Golgi-staining. RESULTS: NPN2 deficient mice had shorter seizure latencies, increased vulnerability to seizure-related death, were more likely to develop spontaneous recurrent seizure activity after chemical challenge, and had an increased slope on input/output curves. Principal cell counts were unchanged, but GABA, parvalbumin, and neuropeptide Y interneuron cell counts were significantly reduced. Synaptosomal NPN2 protein levels and total number of GABAergic synapses were decreased in a gene dose-dependent fashion. CA1 pyramidal cells showed reduced dendritic length and complexity, as well as an increased number of dendritic spines. DISCUSSION: These data suggest the novel hypothesis that the Sema 3F signaling system's role in appropriate placement of subsets of hippocampal interneurons has critical downstream consequences for hippocampal function, resulting in a more seizure susceptible phenotype.


Assuntos
Interneurônios/fisiologia , Neuropilina-2/deficiência , Convulsões/patologia , Convulsões/fisiopatologia , Análise de Variância , Animais , Biofísica , Contagem de Células/métodos , Estimulação Elétrica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Hipocampo/patologia , Técnicas In Vitro , Interneurônios/efeitos dos fármacos , Interneurônios/ultraestrutura , Ácido Caínico , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/fisiopatologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neuropilina-2/genética , Técnicas de Patch-Clamp/métodos , Pentilenotetrazol , Receptores de GABA/metabolismo , Convulsões/induzido quimicamente , Convulsões/genética , Coloração pela Prata/métodos , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo
15.
Neurobiol Aging ; 30(12): 2053-64, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18367293

RESUMO

At hippocampal synapses, repetitive synaptic stimulation (RSS) in the theta frequency range (3-12Hz) is associated with robust EPSP frequency facilitation (FF) and consequently, enhanced action potential (spike) generation and throughput. A complex, synaptically induced hyperpolarization (SIHP) is also triggered by synaptic activation, and a Ca(2+)-dependent afterhyperpolarization (AHP) is triggered above spike threshold. With aging, the AHP is increased and impairs intracellular spike generation, at least in accommodation protocols. However, little is known about how these aging changes interact to affect spike generation at physiological frequencies of RSS, or if the SIHP also is modified in aging. Here we performed the first tests of the net impact of these excitatory and inhibitory aging changes on spike generation during RSS. We report that during RSS at spike threshold (1) spike throughput is well sustained at theta frequencies in young and aged neurons; (2) an interposed AHP dampens spike generation, particularly in aged neurons and at higher frequencies; (3) compared to the AHP, the SIHP does not exert an equivalent inhibitory effect on spike throughput; and (4) in contrast to the AHP, the SIHP is reduced with aging. Together, these results are consistent with a model in which the source of the hyperpolarization is important in determining hippocampal spike throughput within the theta frequency range.


Assuntos
Potenciais de Ação/fisiologia , Envelhecimento/fisiologia , Hipocampo/fisiologia , Potenciais da Membrana/fisiologia , Neurônios/fisiologia , Animais , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas In Vitro , Masculino , Microeletrodos , Modelos Neurológicos , Plasticidade Neuronal/fisiologia , Ratos , Ratos Endogâmicos F344 , Sinapses/fisiologia , Ritmo Teta
16.
Aging Cell ; 6(3): 307-17, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17465978

RESUMO

Evidence accumulated over more than two decades has implicated Ca2+ dysregulation in brain aging and Alzheimer's disease (AD), giving rise to the Ca2+ hypothesis of brain aging and dementia. Electrophysiological, imaging, and behavioral studies in hippocampal or cortical neurons of rodents and rabbits have revealed aging-related increases in the slow afterhyperpolarization, Ca2+ spikes and currents, Ca2+transients, and L-type voltage-gated Ca2+ channel (L-VGCC) activity. Several of these changes have been associated with age-related deficits in learning or memory. Consequently, one version of the Ca2+ hypothesis has been that increased L-VGCC activity drives many of the other Ca2+-related biomarkers of hippocampal aging. In addition, other studies have reported aging- or AD model-related alterations in Ca2+ release from ryanodine receptors (RyR) on intracellular stores. The Ca2+-sensitive RyR channels amplify plasmalemmal Ca2+ influx by the mechanism of Ca2+-induced Ca2+ release (CICR). Considerable evidence indicates that a preferred functional link is present between L-VGCCs and RyRs which operate in series in heart and some brain cells. Here, we review studies implicating RyRs in altered Ca+ regulation in cell toxicity, aging, and AD. A recent study from our laboratory showed that increased CICR plays a necessary role in the emergence of Ca2+-related biomarkers of aging. Consequently, we propose an expanded L-VGCC/Ca2+ hypothesis, in which aging/pathological changes occur in both L-type Ca2+ channels and RyRs, and interact to abnormally amplify Ca2+ transients. In turn, the increased transients result in dysregulation of multiple Ca2+-dependent processes and, through somewhat different pathways, in accelerated functional decline during aging and AD.


Assuntos
Envelhecimento , Doença de Alzheimer/metabolismo , Encéfalo/patologia , Sinalização do Cálcio , Cálcio/metabolismo , Idoso , Animais , Biomarcadores/química , Canais de Cálcio Tipo L/metabolismo , Eletrofisiologia , Humanos , Isquemia , Modelos Biológicos , Neurônios/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
17.
J Neurosci ; 26(13): 3482-90, 2006 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-16571755

RESUMO

Age-dependent changes in multiple Ca2+-related electrophysiological processes in the hippocampus appear to be consistent biomarkers of aging, and several also correlate with cognitive decline. These findings have led to the hypothesis that a common mechanism of Ca2+ dyshomeostasis underlies aspects of aging-dependent brain impairment. However, some key predictions of this view remain untested, including that multiple Ca2+-related biomarkers should emerge concurrently during aging and their onset should also precede/coincide with initial signs of cognitive decline. Moreover, blocking a putative common source of dysregulated Ca2+ should eliminate aging differences. Here, we tested these predictions using combined electrophysiological, imaging, and pharmacological approaches in CA1 neurons to determine the ages of onset (across 4-, 10-, 12-, 14-, and 23-month-old F344 rats) of several established biomarkers, including the increases in the slow afterhyperpolarization, spike accommodation, and [Ca2+]i rise during repetitive synaptic stimulation. In addition, we tested the hypothesis that altered Ca2+-induced Ca2+ release (CICR) from ryanodine receptors, which can be triggered by L-type Ca2+ channels, provides a common source of dysregulated Ca2+ in aging. Results showed that multiple aging biomarkers were first detectable at about the same age (12 months of age; approximately midlife), sufficiently early to influence initial cognitive decline. Furthermore, selectively blocking CICR with ryanodine slowed the Ca2+ rise during synaptic stimulation more in aged rat neurons and, notably, reduced or eliminated aging differences in the biomarkers. Thus, this study provides the first evidence that altered CICR plays a role in driving the early and simultaneous emergence in hippocampus of multiple Ca2+-related biomarkers of aging.


Assuntos
Envelhecimento/fisiologia , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Hipocampo/fisiologia , Neurônios/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Homeostase/fisiologia , Ativação do Canal Iônico , Masculino , Ratos , Ratos Endogâmicos F344 , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...