Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Aging Cell ; 19(1): e13057, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31693798

RESUMO

Cerebrovascular dysfunction and cognitive decline are highly prevalent in aging, but the mechanisms underlying these impairments are unclear. Cerebral blood flow decreases with aging and is one of the earliest events in the pathogenesis of Alzheimer's disease (AD). We have previously shown that the mechanistic/mammalian target of rapamycin (mTOR) drives disease progression in mouse models of AD and in models of cognitive impairment associated with atherosclerosis, closely recapitulating vascular cognitive impairment. In the present studies, we sought to determine whether mTOR plays a role in cerebrovascular dysfunction and cognitive decline during normative aging in rats. Using behavioral tools and MRI-based functional imaging, together with biochemical and immunohistochemical approaches, we demonstrate that chronic mTOR attenuation with rapamycin ameliorates deficits in learning and memory, prevents neurovascular uncoupling, and restores cerebral perfusion in aged rats. Additionally, morphometric and biochemical analyses of hippocampus and cortex revealed that mTOR drives age-related declines in synaptic and vascular density during aging. These data indicate that in addition to mediating AD-like cognitive and cerebrovascular deficits in models of AD and atherosclerosis, mTOR drives cerebrovascular, neuronal, and cognitive deficits associated with normative aging. Thus, inhibitors of mTOR may have potential to treat age-related cerebrovascular dysfunction and cognitive decline. Since treatment of age-related cerebrovascular dysfunction in older adults is expected to prevent further deterioration of cerebral perfusion, recently identified as a biomarker for the very early (preclinical) stages of AD, mTOR attenuation may potentially block the initiation and progression of AD.


Assuntos
Envelhecimento/genética , Circulação Cerebrovascular/fisiologia , Disfunção Cognitiva/fisiopatologia , Serina-Treonina Quinases TOR/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Ratos
2.
J Alzheimers Dis ; 64(s1): S427-S453, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29710715

RESUMO

Insulin resistance can occur when the body is unable to respond to insulin even in excess. In the brain, insulin manages glucose metabolism in regions such as the hippocampus and plays a key role in directly regulating ERK, a kinase required for the type of memory compromised in early Alzheimer's disease (AD). Human imaging studies show that brain glucose utilization declines with age and is notably impaired in subjects with early AD. Likewise, animal models of AD or insulin resistance, or both, demonstrate that dysfunctional insulin signaling and insulin resistance in the brain have reciprocity with neuroinflammation and aberrant accumulation of amyloid-ß (Aß), pathological hallmarks in AD. As such, the association between brain insulin activity and AD has led to clinical trials testing the efficacy of insulin and insulin-sensitizing drugs to intervene in AD. Based on recent inquiries to ClinicalTrials.gov, we evaluated thirty-three clinical studies related to AD and insulin. The search filtered for interventional clinical trials to test FDA-approved drugs or substances that impinge upon the insulin signaling pathway. Insulin, metformin, and thiazolidinediones were the three main interventions assessed. Overall, these strategies are expected to negate the effects of brain insulin resistance by targeting insulin signaling pathways involved in neuroinflammation, metabolic homeostasis, synaptic functional and structural integrity. The goal of this review is to provide an update on insulin and ERK signaling in relation to memory, its decline in early AD, and provide an overview of clinical trials related to insulin for early AD intervention.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Insulina/metabolismo , Animais , Humanos
3.
Am J Physiol Heart Circ Physiol ; 314(4): H693-H703, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351469

RESUMO

An intact blood-brain barrier (BBB) limits entry of proinflammatory and neurotoxic blood-derived factors into the brain parenchyma. The BBB is damaged in Alzheimer's disease (AD), which contributes significantly to the progression of AD pathologies and cognitive decline. However, the mechanisms underlying BBB breakdown in AD remain elusive, and no interventions are available for treatment or prevention. We and others recently established that inhibition of the mammalian/mechanistic target of rapamycin (mTOR) pathway with rapamycin yields significant neuroprotective effects, improving cerebrovascular and cognitive function in mouse models of AD. To test whether mTOR inhibition protects the BBB in neurological diseases of aging, we treated hAPP(J20) mice modeling AD and low-density lipoprotein receptor-null (LDLR-/-) mice modeling vascular cognitive impairment with rapamycin. We found that inhibition of mTOR abrogates BBB breakdown in hAPP(J20) and LDLR-/- mice. Experiments using an in vitro BBB model indicated that mTOR attenuation preserves BBB integrity through upregulation of specific tight junction proteins and downregulation of matrix metalloproteinase-9 activity. Together, our data establish mTOR activity as a critical mediator of BBB breakdown in AD and, potentially, vascular cognitive impairment and suggest that rapamycin and/or rapalogs could be used for the restoration of BBB integrity. NEW & NOTEWORTHY This report establishes mammalian/mechanistic target of rapamycin as a critical mediator of blood-brain barrier breakdown in models of Alzheimer's disease and vascular cognitive impairment and suggests that drugs targeting the target of rapamycin pathway could be used for the restoration of blood-brain barrier integrity in disease states.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Comportamento Animal , Barreira Hematoencefálica/efeitos dos fármacos , Cognição , Demência Vascular/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Animais , Barreira Hematoencefálica/enzimologia , Barreira Hematoencefálica/patologia , Linhagem Celular , Demência Vascular/enzimologia , Demência Vascular/patologia , Demência Vascular/psicologia , Modelos Animais de Doenças , Feminino , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/deficiência , Receptores de LDL/genética , Serina-Treonina Quinases TOR/metabolismo , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/enzimologia , Junções Íntimas/patologia
4.
J Cereb Blood Flow Metab ; 38(1): 58-74, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28511572

RESUMO

We recently showed that mTOR attenuation blocks progression and abrogates established cognitive deficits in Alzheimer's disease (AD) mouse models. These outcomes were associated with the restoration of cerebral blood flow (CBF) and brain vascular density (BVD) resulting from relief of mTOR inhibition of NO release. Recent reports suggested a role of mTOR in atherosclerosis. Because mTOR drives aging and vascular dysfunction is a universal feature of aging, we hypothesized that mTOR may contribute to brain vascular and cognitive dysfunction associated with atherosclerosis. We measured CBF, BVD, cognitive function, markers of inflammation, and parameters of cardiovascular disease in LDLR-/- mice fed maintenance or high-fat diet ± rapamycin. Cardiovascular pathologies were proportional to severity of brain vascular dysfunction. Aortic atheromas were reduced, CBF and BVD were restored, and cognitive dysfunction was attenuated potentially through reduction in systemic and brain inflammation following chronic mTOR attenuation. Our studies suggest that mTOR regulates vascular integrity and function and that mTOR attenuation may restore neurovascular function and cardiovascular health. Together with our previous studies in AD models, our data suggest mTOR-driven vascular damage may be a mechanism shared by age-associated neurological diseases. Therefore, mTOR attenuation may have promise for treatment of cognitive impairment in atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Circulação Cerebrovascular/fisiologia , Disfunção Cognitiva/metabolismo , Demência Vascular/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Aterosclerose/complicações , Disfunção Cognitiva/etiologia , Demência Vascular/etiologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Receptores de LDL/deficiência
5.
Aging Dis ; 8(3): 257-266, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28580182

RESUMO

The importance of vascular contributions to cognitive impairment and dementia (VCID) associated with Alzheimer's disease (AD) and related neurodegenerative diseases is increasingly recognized, however, the underlying mechanisms remain obscure. There is growing evidence that in addition to Aß deposition, accumulation of hyperphosphorylated oligomeric tau contributes significantly to AD etiology. Tau oligomers are toxic and it has been suggested that they propagate in a "prion-like" fashion, inducing endogenous tau misfolding in cells. Their role in VCID, however, is not yet understood. The present study was designed to determine the severity of vascular deposition of oligomeric tau in the brain in patients with AD and related tauopathies, including dementia with Lewy bodies (DLB) and progressive supranuclear palsy (PSP). Further, we examined a potential link between vascular deposition of fibrillar Aß and that of tau oligomers in the Tg2576 mouse model. We found that tau oligomers accumulate in cerebral microvasculature of human patients with AD and PSP, in association with vascular endothelial and smooth muscle cells. Cerebrovascular deposition of tau oligomers was also found in DLB patients. We also show that tau oligomers accumulate in cerebral microvasculature of Tg2576 mice, partially in association with cerebrovascular Aß deposits. Thus, our findings add to the growing evidence for multifaceted microvascular involvement in the pathogenesis of AD and other neurodegenerative diseases. Accumulation of tau oligomers may represent a potential novel mechanism by which functional and structural integrity of the cerebral microvessels is compromised.

6.
J Cereb Blood Flow Metab ; 37(1): 217-226, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26721390

RESUMO

Apolipoprotein E ɛ4 allele is a common susceptibility gene for late-onset Alzheimer's disease. Brain vascular and metabolic deficits can occur in cognitively normal apolipoprotein E ɛ4 carriers decades before the onset of Alzheimer's disease. The goal of this study was to determine whether early intervention using rapamycin could restore neurovascular and neurometabolic functions, and thus impede pathological progression of Alzheimer's disease-like symptoms in pre-symptomatic Apolipoprotein E ɛ4 transgenic mice. Using in vivo, multimodal neuroimaging, we found that apolipoprotein E ɛ4 mice treated with rapamycin had restored cerebral blood flow, blood-brain barrier integrity and glucose metabolism, compared to age- and gender-matched wild-type controls. The preserved vasculature and metabolism were associated with amelioration of incipient learning deficits. We also found that rapamycin restored the levels of the proinflammatory cyclophilin A in vasculature, which may contribute to the preservation of cerebrovascular function in the apolipoprotein E ɛ4 transgenics. Our results show that rapamycin improves functional outcomes in this mouse model and may have potential as an effective intervention to block progression of vascular, metabolic and early cognitive deficits in human Apolipoprotein E ɛ4 carriers. As rapamycin is FDA-approved and neuroimaging is readily used in humans, the results of the present study may provide the basis for future Alzheimer's disease intervention studies in human subjects.


Assuntos
Doença de Alzheimer/patologia , Apolipoproteína E4/genética , Prevenção Secundária/métodos , Sirolimo/uso terapêutico , Doença de Alzheimer/complicações , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Transtornos do Metabolismo de Glucose/tratamento farmacológico , Deficiências da Aprendizagem/tratamento farmacológico , Deficiências da Aprendizagem/prevenção & controle , Doenças Metabólicas/tratamento farmacológico , Camundongos , Camundongos Transgênicos , Neuroimagem , Prevenção Secundária/tendências , Sirolimo/farmacologia , Doenças Vasculares/tratamento farmacológico
7.
Curr Top Med Chem ; 15(21): 2139-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26059360

RESUMO

With the global aging population, Alzheimer's disease, Parkinson's disease and mild cognition impairment are increasing in prevalence. The success of rapamycin as an agent to extend lifespan in various organisms, including mice, brings hope that chronic mTOR inhibition could also refrain age-related neurodegeneration. Here we review the evidence suggesting that mTOR inhibition - mainly with rapamycin - is a valid intervention to delay age-related neurodegeneration. We discuss the potential mechanisms by which rapamycin may facilitate neurodegeneration prevention or restoration of cognitive function. We also discuss the known side effects of rapamycin and provide evidence to alleviate exaggerated concerns regarding its wider clinical use. We explore the small molecule alternatives to rapamycin and propose future directions for their development, mainly by exploring the possibility of targeting the downstream effectors of mTOR: S6K1 and especially S6K2. Finally, we discuss the strengths and weaknesses of the models used to determine intervention efficacy for neurodegeneration. We address the difficulties of interpreting data using the common way of investigating the efficacy of interventions to delay/prevent neurodegeneration by observing animal behavior while these animals are under treatment. We propose an experimental design that should isolate the variable of aging in the experimental design and resolve the ambiguity present in recent literature.


Assuntos
Terapia de Alvo Molecular/métodos , Doenças Neurodegenerativas/prevenção & controle , Serina-Treonina Quinases TOR/antagonistas & inibidores , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Animais , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Camundongos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Sirolimo/efeitos adversos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
8.
Neurobiol Dis ; 72 Pt A: 92-103, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25237037

RESUMO

Insulin is a key hormone regulating metabolism. Insulin binding to cell surface insulin receptors engages many signaling intermediates operating in parallel and in series to control glucose, energy, and lipids while also regulating mitogenesis and development. Perturbations in the function of any of these intermediates, which occur in a variety of diseases, cause reduced sensitivity to insulin and insulin resistance with consequent metabolic dysfunction. Chronic inflammation ensues which exacerbates compromised metabolic homeostasis. Since insulin has a key role in learning and memory as well as directly regulating ERK, a kinase required for the type of learning and memory compromised in early Alzheimer's disease (AD), insulin resistance has been identified as a major risk factor for the onset of AD. Animal models of AD or insulin resistance or both demonstrate that AD pathology and impaired insulin signaling form a reciprocal relationship. Of note are human and animal model studies geared toward improving insulin resistance that have led to the identification of the nuclear receptor and transcription factor, peroxisome proliferator-activated receptor gamma (PPARγ) as an intervention tool for early AD. Strategic targeting of alternate nodes within the insulin signaling network has revealed disease-stage therapeutic windows in animal models that coalesce with previous and ongoing clinical trial approaches. Thus, exploiting the connection between insulin resistance and AD provides powerful opportunities to delineate therapeutic interventions that slow or block the pathogenesis of AD.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Resistência à Insulina , Insulina/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Encefalite/metabolismo , Humanos , Aprendizagem/fisiologia , Sistema de Sinalização das MAP Quinases , PPAR gama/metabolismo
9.
J Neurosci ; 34(11): 4054-63, 2014 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-24623782

RESUMO

Cognitive impairment is a quintessential feature of Alzheimer's disease (AD) and AD mouse models. The peroxisome proliferator-activated receptor-γ (PPARγ) agonist rosiglitazone improves hippocampus-dependent cognitive deficits in some AD patients and ameliorates deficits in the Tg2576 mouse model for AD amyloidosis. Tg2576 cognitive enhancement occurs through the induction of a gene and protein expression profile reflecting convergence of the PPARγ signaling axis and the extracellular signal-regulated protein kinase (ERK) cascade, a critical mediator of memory consolidation. We therefore tested whether PPARγ and ERK associated in protein complexes that subserve cognitive enhancement through PPARγ agonism. Coimmunoprecipitation of hippocampal extracts revealed that PPARγ and activated, phosphorylated ERK (pERK) associated in Tg2576 in vivo, and that PPARγ agonism facilitated recruitment of PPARγ to pERK during memory consolidation. Furthermore, the amount of PPARγ recruited to pERK correlated with the cognitive reserve in humans with AD and in Tg2576. Our findings implicate a previously unidentified PPARγ-pERK complex that provides a molecular mechanism for the convergence of these pathways during cognitive enhancement, thereby offering new targets for therapeutic development in AD.


Assuntos
Doença de Alzheimer/metabolismo , Cognição/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Memória/fisiologia , PPAR gama/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Anilidas/farmacologia , Animais , Cognição/efeitos dos fármacos , Feminino , Hipocampo/fisiologia , Humanos , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Pessoa de Meia-Idade , Nootrópicos/farmacologia , PPAR gama/antagonistas & inibidores , Fosforilação/fisiologia , Rosiglitazona , Tiazolidinedionas/farmacologia
10.
Curr Trends Neurol ; 8: 31-38, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-34924724

RESUMO

Increasing evidence suggests that vascular dysfunction, a universal feature of aging, mechanistically contributes to the onset and pathogenesis of neurological diseases of aging. It was recently discovered that attenuating activity of the mammalian/mechanistic target of rapamycin (mTOR) extends both life- and health-span in mice by delaying aging. Here we review current evidence for a critical role of mTOR in age-associated vascular dysfunction and discuss potential mechanisms by which this pathway may lead to cognitive decline in Alzheimer's disease.

11.
J Neurosci ; 32(47): 16725-35a, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23175826

RESUMO

We previously reported that the peroxisome proliferator-activated receptor γ (PPARγ) agonist rosiglitazone (RSG) improved hippocampus-dependent cognition in the Alzheimer's disease (AD) mouse model, Tg2576. RSG had no effect on wild-type littermate cognitive performance. Since extracellular signal-regulated protein kinase mitogen-activated protein kinase (ERK MAPK) is required for many forms of learning and memory that are affected in AD, and since both PPARγ and ERK MAPK are key mediators of insulin signaling, the current study tested the hypothesis that RSG-mediated cognitive improvement induces a hippocampal PPARγ pattern of gene and protein expression that converges with the ERK MAPK signaling axis in Tg2576 AD mice. In the hippocampal PPARγ transcriptome, we found significant overlap between peroxisome proliferator response element-containing PPARγ target genes and ERK-regulated, cAMP response element-containing target genes. Within the Tg2576 dentate gyrus proteome, RSG induced proteins with structural, energy, biosynthesis and plasticity functions. Several of these proteins are known to be important for cognitive function and are also regulated by ERK MAPK. In addition, we found the RSG-mediated augmentation of PPARγ and ERK2 activity during Tg2576 cognitive enhancement was reversed when hippocampal PPARγ was pharmacologically antagonized, revealing a coordinate relationship between PPARγ transcriptional competency and phosphorylated ERK that is reciprocally affected in response to chronic activation, compared with acute inhibition, of PPARγ. We conclude that the hippocampal transcriptome and proteome induced by cognitive enhancement with RSG harnesses a dysregulated ERK MAPK signal transduction pathway to overcome AD-like cognitive deficits in Tg2576 mice. Thus, PPARγ represents a signaling system that is not crucial for normal cognition yet can intercede to restore neural networks compromised by AD.


Assuntos
Hipocampo/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Nootrópicos/farmacologia , PPAR gama/fisiologia , Transdução de Sinais/fisiologia , Tiazolidinedionas/farmacologia , Peptídeos beta-Amiloides/metabolismo , Animais , Western Blotting , Núcleo Celular/fisiologia , Condicionamento Psicológico , Eletrochoque , Medo , Feminino , Injeções Intraventriculares , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/antagonistas & inibidores , Reação em Cadeia da Polimerase , Rosiglitazona , Espectrometria de Massas em Tandem , Transcriptoma/fisiologia
12.
Am J Trop Med Hyg ; 82(5): 954-60, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20439981

RESUMO

Viral hemorrhagic fever is caused by a diverse group of single-stranded, negative-sense or positive-sense RNA viruses belonging to the families Filoviridae (Ebola and Marburg), Arenaviridae (Lassa, Junin, Machupo, Sabia, and Guanarito), and Bunyaviridae (hantavirus). Disease characteristics in these families mark each with the potential to be used as a biological threat agent. Because other diseases have similar clinical symptoms, specific laboratory diagnostic tests are necessary to provide the differential diagnosis during outbreaks and for instituting acceptable quarantine procedures. We designed 48 TaqMan-based polymerase chain reaction (PCR) assays for specific and absolute quantitative detection of multiple hemorrhagic fever viruses. Forty-six assays were determined to be virus-specific, and two were designated as pan assays for Marburg virus. The limit of detection for the assays ranged from 10 to 0.001 plaque-forming units (PFU)/PCR. Although these real-time hemorrhagic fever virus assays are qualitative (presence of target), they are also quantitative (measure a single DNA/RNA target sequence in an unknown sample and express the final results as an absolute value (e.g., viral load, PFUs, or copies/mL) on the basis of concentration of standard samples and can be used in viral load, vaccine, and antiviral drug studies.


Assuntos
Arenavirus/isolamento & purificação , Filoviridae/isolamento & purificação , Orthohantavírus/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , Arenavirus/classificação , Arenavirus/genética , Filoviridae/classificação , Filoviridae/genética , Orthohantavírus/classificação , Orthohantavírus/genética , Humanos , RNA Viral/classificação , RNA Viral/isolamento & purificação , Sensibilidade e Especificidade
13.
Am J Trop Med Hyg ; 81(4): 679-84, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19815886

RESUMO

Chikungunya (CHIK) and O'nyong-nyong (ONN) are important emerging arthropod-borne diseases. Molecular diagnosis of these two viruses in mosquitoes has not been evaluated, and the effects of extraneous mosquito tissue on assay performance have not been tested. Additionally, no real-time reverse transcription-polymerase chain reaction (RT-PCR) assay exists for detecting ONN virus (ONNV) RNA. We describe the development of sensitive and specific real-time RT-PCR assays for detecting CHIK and ONN viral RNA in mosquitoes, which have application for field use. In addition, we compared three methods for primer/probe design for assay development by evaluating their sensitivity and specificity. This comparison resulted in development of virus-specific assays that could detect less than one plaque-forming unit equivalent of each of the viruses in mosquitoes. The use of these assays will aid in arthropod-borne disease surveillance and in the control of the associated diseases.


Assuntos
Vírus Chikungunya/isolamento & purificação , Culicidae/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Animais , RNA Viral/isolamento & purificação , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...