Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Glia ; 69(9): 2059-2076, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33638562

RESUMO

Gliomas are the most common primary intrinsic brain tumors occurring in adults. Of all malignant gliomas, glioblastoma (GBM) is considered the deadliest tumor type due to diffuse brain invasion, immune evasion, cellular, and molecular heterogeneity, and resistance to treatments resulting in high rates of recurrence. An extensive understanding of the genomic and microenvironmental landscape of gliomas gathered over the past decade has renewed interest in pursuing novel therapeutics, including immune checkpoint inhibitors, glioma-associated macrophage/microglia (GAMs) modulators, and others. In light of this, predictive animal models that closely recreate the conditions and findings found in human gliomas will serve an increasingly important role in identifying new, effective therapeutic strategies. Although numerous syngeneic, xenograft, and transgenic rodent models have been developed, few include the full complement of pathobiological features found in human tumors, and therefore few accurately predict bench-to-bedside success. This review provides an update on how genetically engineered rodent models based on the replication-competent avian-like sarcoma (RCAS) virus/tumor virus receptor-A (tv-a) system have been used to recapitulate key elements of human gliomas in an immunologically intact host microenvironment and highlights new approaches using this model system as a predictive tool for advancing translational glioma research.


Assuntos
Neoplasias Encefálicas , Modelos Animais de Doenças , Glioma , Sarcoma , Animais , Vírus do Sarcoma Aviário/genética , Neoplasias Encefálicas/patologia , Glioma/patologia , Humanos , Vírus Oncogênicos , Receptores Virais , Microambiente Tumoral
2.
Glia ; 69(9): 2199-2214, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33991013

RESUMO

High-grade gliomas (HGGs) are aggressive, treatment-resistant, and often fatal human brain cancers. The TNF-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor-inducible 14 (Fn14) signaling axis is involved in tissue repair after injury and constitutive signaling has been implicated in the pathogenesis of numerous solid cancers. The Fn14 gene is expressed at low levels in the normal, uninjured brain but is highly expressed in primary isocitrate dehydrogenase wild-type and recurrent HGGs. Fn14 signaling is implicated in numerous aspects of glioma biology including brain invasion and chemotherapy resistance, but whether Fn14 overexpression can directly promote tumor malignancy has not been reported. Here, we used the replication-competent avian sarcoma-leukosis virus/tumor virus A system to examine the impact of Fn14 expression on glioma development and pathobiology. We found that the sole addition of Fn14 to an established oncogenic cocktail previously shown to generate proneural-like gliomas led to the development of highly invasive and lethal brain cancer with striking biological features including extensive pseudopalisading necrosis, constitutive canonical and noncanonical NF-κB pathway signaling, and high plasminogen activator inhibitor-1 (PAI-1) expression. Analyses of HGG patient datasets revealed that high human PAI-1 gene (SERPINE1) expression correlates with shorter patient survival, and that the SERPINE1 and Fn14 (TNFRSF12A) genes are frequently co-expressed in bulk tumor tissues, in tumor subregions, and in malignant cells residing in the tumor microenvironment. These findings provide new insights into the potential importance of Fn14 in human HGG pathobiology and designate both the NF-κB signaling node and PAI-1 as potential targets for therapeutic intervention. MAIN POINTS: This work demonstrates that elevated levels of the TWEAK receptor Fn14 in tumor-initiating, neural progenitor cells leads to the transformation of proneural-like gliomas into more aggressive and lethal tumors that exhibit constitutive NF-κB pathway activation and plasminogen activator inhibitor-1 overexpression.


Assuntos
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Fatores de Crescimento de Fibroblastos , Glioma/patologia , Humanos , Invasividade Neoplásica , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor de TWEAK , Microambiente Tumoral
3.
Arch Ital Biol ; 150(1): 5-14, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22786833

RESUMO

Prior research has reported beneficial effects of melatonin in rodent models of Alzheimer's disease (AD). This study evaluated the effect of ramelteon (Rozerem, a melatonin receptor agonist) on spatial learning & memory and neuropathological markers in a transgenic murine model of AD (the B6C3-Tg(APPswe,PSEN1dE9)85Dbo/J transgenic mouse strain; hereafter 'AD mice'). Three months of daily ramelteon treatment (~3mg/kg/day), starting at 3 months of age, did not produce an improvement in the cognitive performance of AD mice (water maze). In contrast to wild-type control mice, AD mice did not show any evidence of having learned the location of the escape platform. The cortex and hippocampus of AD mice contained significant quantities of beta-amyloid plaques and PARP-positive (poly ADP ribose polymerase) cells, indicating apoptosis. Six months of ramelteon treatment, starting at 3 months of age, did not produce any change in these neuropathological markers. The ability of long term melatonin treatment to improve cognition and attenuate neuropathology in AD mice did not generalize to this dosage of ramelteon.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Antipsicóticos/uso terapêutico , Encéfalo/metabolismo , Indenos/uso terapêutico , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Apoptose/genética , Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Seguimentos , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Mutação/genética , Placa Amiloide/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Presenilina-1/genética , Fatores de Tempo
4.
Sci Adv ; 6(3): eaax3931, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31998833

RESUMO

Development of effective tumor cell-targeted nanodrug formulations has been quite challenging, as many nanocarriers and targeting moieties exhibit nonspecific binding to cellular, extracellular, and intravascular components. We have developed a therapeutic nanoparticle formulation approach that balances cell surface receptor-specific binding affinity while maintaining minimal interactions with blood and tumor tissue components (termed "DART" nanoparticles), thereby improving blood circulation time, biodistribution, and tumor cell-specific uptake. Here, we report that paclitaxel (PTX)-DART nanoparticles directed to the cell surface receptor fibroblast growth factor-inducible 14 (Fn14) outperformed both the corresponding PTX-loaded, nontargeted nanoparticles and Abraxane, an FDA-approved PTX nanoformulation, in both a primary triple-negative breast cancer (TNBC) model and an intracranial model reflecting TNBC growth following metastatic dissemination to the brain. These results provide new insights into methods for effective development of therapeutic nanoparticles as well as support the continued development of the DART platform for primary and metastatic tumors.


Assuntos
Antineoplásicos/administração & dosagem , Biomarcadores Tumorais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Terapia de Alvo Molecular , Nanopartículas , Nanomedicina Teranóstica , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/mortalidade , Modelos Animais de Doenças , Matriz Extracelular , Feminino , Expressão Gênica , Humanos , Camundongos , Terapia de Alvo Molecular/efeitos adversos , Terapia de Alvo Molecular/métodos , Metástase Neoplásica , Estadiamento de Neoplasias , Prognóstico , RNA Mensageiro , Receptor de TWEAK/genética , Distribuição Tecidual , Resultado do Tratamento , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/etiologia , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Photochem Photobiol ; 96(2): 301-309, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31441057

RESUMO

Fluorescence-guided surgery (FGS) is routinely utilized in clinical centers around the world, whereas the combination of FGS and photodynamic therapy (PDT) has yet to reach clinical implementation and remains an active area of translational investigations. Two significant challenges to the clinical translation of PDT for brain cancer are as follows: (1) Limited light penetration depth in brain tissues and (2) Poor selectivity and delivery of the appropriate photosensitizers. To address these shortcomings, we developed nanoliposomal protoporphyrin IX (Nal-PpIX) and nanoliposomal benzoporphyrin derivative (Nal-BPD) and then evaluated their photodynamic effects as a function of depth in tissue and light fluence using rat brains. Although red light penetration depth (defined as the depth at which the incident optical energy drops to 1/e, ~37%) is typically a few millimeters in tissues, we demonstrated that the remaining optical energy could induce PDT effects up to 2 cm within brain tissues. Photobleaching and singlet oxygen yield studies between Nal-BPD and Nal-PpIX suggest that deep-tissue PDT (>1 cm) is more effective when using Nal-BPD. These findings indicate that Nal-BPD-PDT is more likely to generate cytotoxic effects deep within the brain and allow for the treatment of brain invading tumor cells centimeters away from the main, resectable tumor mass.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacocinética , Animais , Neoplasias Encefálicas/metabolismo , Fármacos Fotossensibilizantes/uso terapêutico , Ratos , Espectrometria de Fluorescência
6.
J Sleep Res ; 17(4): 376-84, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19021853

RESUMO

A novel animal-analog of the human psychomotor vigilance task (PVT) was validated by subjecting rats to 24 h of sleep deprivation (SD) and examining the effect on performance in the rat-PVT (rPVT), and a rat multiple sleep latency test (rMSLT). During a three-phase (separate cohorts) crossover design, vigilance performance in the rPVT was compared with 24 h SD-induced changes in sleepiness assessed by polysomnographic evaluation and the rMSLT. Twenty-four hours of SD was produced by brief rotation of activity wheels at regular intervals in which the animals resided throughout the experiment. In the rPVT experiment, exercise controls (EC) experienced the same overall amount of locomotor activity as during SD, but allowed long periods of undisturbed sleep. After 24 h SD response latencies slowed, and lapses increased significantly during rPVT performance when compared with baseline and EC conditions. During the first 3 h of the recovery period following 24 h SD, polysomnographic measures indicated sleepiness. Latency to fall asleep after 24 h SD was assessed six times during the first 3 h after SD. Rats fell asleep significantly faster immediately after SD, than after non-SD baseline sessions. In conclusion, 24 h of SD in rats increased sleepiness, as indicated by polysomnography and the rMSLT, and impaired vigilance as measured by the rPVT. The rPVT closely resembles the human PVT test widely used in human sleep research and will assist investigation of the neurobiologic mechanisms that produce vigilance impairments after sleep disruption.


Assuntos
Nível de Alerta/fisiologia , Distúrbios do Sono por Sonolência Excessiva/epidemiologia , Distúrbios do Sono por Sonolência Excessiva/etiologia , Desempenho Psicomotor , Privação do Sono/complicações , Privação do Sono/epidemiologia , Animais , Distúrbios do Sono por Sonolência Excessiva/diagnóstico , Eletroencefalografia , Eletromiografia , Locomoção , Polissonografia , Ratos , Ratos Endogâmicos F344 , Tempo de Reação , Índice de Gravidade de Doença
7.
Sci Rep ; 8(1): 1180, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29352201

RESUMO

Glioma is a unique neoplastic disease that develops exclusively in the central nervous system (CNS) and rarely metastasizes to other tissues. This feature strongly implicates the tumor-host CNS microenvironment in gliomagenesis and tumor progression. We investigated the differences and similarities in glioma biology as conveyed by transcriptomic patterns across four mammalian hosts: rats, mice, dogs, and humans. Given the inherent intra-tumoral molecular heterogeneity of human glioma, we focused this study on tumors with upregulation of the platelet-derived growth factor signaling axis, a common and early alteration in human gliomagenesis. The results reveal core neoplastic alterations in mammalian glioma, as well as unique contributions of the tumor host to neoplastic processes. Notable differences were observed in gene expression patterns as well as related biological pathways and cell populations known to mediate key elements of glioma biology, including angiogenesis, immune evasion, and brain invasion. These data provide new insights regarding mammalian models of human glioma, and how these insights and models relate to our current understanding of the human disease.


Assuntos
Neoplasias Encefálicas/genética , Transformação Celular Neoplásica/genética , Perfilação da Expressão Gênica , Glioma/genética , Transcriptoma , Animais , Neoplasias Encefálicas/patologia , Biologia Computacional/métodos , Cães , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Camundongos , Ratos , Reprodutibilidade dos Testes , Especificidade da Espécie
8.
J Control Release ; 267: 144-153, 2017 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-28887134

RESUMO

The most common and deadly form of primary brain cancer, glioblastoma (GBM), is characterized by significant intratumoral heterogeneity, microvascular proliferation, immune system suppression, and brain tissue invasion. Delivering effective and sustained treatments to the invasive GBM cells intermixed with functioning neural elements is a major goal of advanced therapeutic systems for brain cancer. Previously, we investigated the nanoparticle characteristics that enable targeting of invasive GBM cells. This revealed the importance of minimizing non-specific binding within the relatively adhesive, 'sticky' microenvironment of the brain and brain tumors in particular. We refer to such nanoformulations with decreased non-specific adhesivity and receptor targeting as 'DART' therapeutics. In this work, we applied this information toward the design and characterization of biodegradable nanocarriers, and in vivo testing in orthotopic experimental gliomas. We formulated particulate nanocarriers using poly(lactic-co-glycolic acid) (PLGA) and PLGA-polyethylene glycol (PLGA-PEG) polymers to generate sub-100nm nanoparticles with minimal binding to extracellular brain components and strong binding to the Fn14 receptor - an upregulated, conserved component in invasive GBM. Multiple particle tracking in brain tissue slices and in vivo testing in orthotopic murine malignant glioma revealed preserved nanoparticle diffusivity and increased uptake in brain tumor cells. These combined characteristics also resulted in longer retention of the DART nanoparticles within the orthotopic tumors compared to non-targeted versions. Taken together, these results and nanoparticle design considerations offer promising new methods to optimize therapeutic nanocarriers for improving drug delivery and treatment for invasive brain tumors.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Portadores de Fármacos/administração & dosagem , Glioma/tratamento farmacológico , Nanopartículas/administração & dosagem , Receptor de TWEAK/metabolismo , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Encéfalo/metabolismo , Linhagem Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Proteínas da Matriz Extracelular/metabolismo , Glioma/metabolismo , Camundongos Endogâmicos C57BL , Nanopartículas/química , Poliésteres/administração & dosagem , Poliésteres/química , Poliésteres/farmacocinética , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética
9.
PLoS One ; 12(3): e0174557, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28358926

RESUMO

Previously rodent preclinical research in gliomas frequently involved implantation of cell lines such as C6 and 9L into the rat brain. More recently, mouse models have taken over, the genetic manipulability of the mouse allowing the creation of genetically accurate models outweighed the disadvantage of its smaller brain size that limited time allowed for tumor progression. Here we illustrate a method that allows glioma formation in the rat using the replication competent avian-like sarcoma (RCAS) virus / tumor virus receptor-A (tv-a) transgenic system of post-natal cell type-specific gene transfer. The RCAS/tv-a model has emerged as a particularly versatile and accurate modeling technology by enabling spatial, temporal, and cell type-specific control of individual gene transformations and providing de novo formed glial tumors with distinct molecular subtypes mirroring human GBM. Nestin promoter-driven tv-a (Ntv-a) transgenic Sprague-Dawley rat founder lines were created and RCAS PDGFA and p53 shRNA constructs were used to initiate intracranial brain tumor formation. Tumor formation and progression were confirmed and visualized by magnetic resonance imaging (MRI) and spectroscopy. The tumors were analyzed using histopathological and immunofluorescent techniques. All experimental animals developed large, heterogeneous brain tumors that closely resembled human GBM. Median survival was 92 days from tumor initiation and 62 days from the first point of tumor visualization on MRI. Each tumor-bearing animal showed time dependent evidence of malignant progression to high-grade glioma by MRI and neurological examination. Post-mortem tumor analysis demonstrated the presence of several key characteristics of human GBM, including high levels of tumor cell proliferation, pseudopalisading necrosis, microvascular proliferation, invasion of tumor cells into surrounding tissues, peri-tumoral reactive astrogliosis, lymphocyte infiltration, presence of numerous tumor-associated microglia- and bone marrow-derived macrophages, and the formation of stem-like cell niches within the tumor. This transgenic rat model may enable detailed interspecies comparisons of fundamental cancer pathways and clinically relevant experimental imaging procedures and interventions that are limited by the smaller size of the mouse brain.


Assuntos
Encéfalo/diagnóstico por imagem , Glioma/genética , Nestina/genética , Fator de Crescimento Derivado de Plaquetas/genética , Proteína Supressora de Tumor p53/genética , Animais , Vírus do Sarcoma Aviário/genética , Vírus do Sarcoma Aviário/patogenicidade , Encéfalo/patologia , Encéfalo/virologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Engenharia Genética , Glioma/diagnóstico por imagem , Glioma/patologia , Glioma/virologia , Humanos , Macrófagos/patologia , Imageamento por Ressonância Magnética , Camundongos , Ratos , Ratos Transgênicos
10.
Curr Pharm Des ; 22(9): 1177-1193, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26685681

RESUMO

The blood-brain barrier (BBB) poses a unique challenge for drug delivery to the central nervous system (CNS). The BBB consists of a continuous layer of specialized endothelial cells linked together by tight junctions, pericytes, nonfenestrated basal lamina, and astrocytic foot processes. This complex barrier controls and limits the systemic delivery of therapeutics to the CNS. Several innovative strategies have been explored to enhance the transport of therapeutics across the BBB, each with individual advantages and disadvantages. Ongoing advances in delivery approaches that overcome the BBB are enabling more effective therapies for CNS diseases. In this review, we discuss: (1) the physiological properties of the BBB, (2) conventional strategies to enhance paracellular and transcellular transport through the BBB, (3) emerging concepts to overcome the BBB, and (4) alternative CNS drug delivery strategies that bypass the BBB entirely. Based on these exciting advances, we anticipate that in the near future, drug delivery research efforts will lead to more effective therapeutic interventions for diseases of the CNS.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Encefalopatias/tratamento farmacológico , Fármacos do Sistema Nervoso Central/uso terapêutico , Sistemas de Liberação de Medicamentos , Animais , Humanos
11.
Front Psychol ; 6: 394, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25904885

RESUMO

The present experiment investigated potential reinstaters of suboptimal economic decision making in rats. Rats were first trained on a version of the rat Gambling Task under conditions designed to promote choice of a suboptimal option that occasionally resulted in large "wins" (four sucrose pellets). In a second phase, preference for this economically suboptimal option was reduced by substantially increasing the probability of punishment when this option was chosen. Then, three events were tested for their ability to reinstate choice of the suboptimal option. A brief period of re-exposure to a high frequency of large wins significantly increased choice of the suboptimal option. The pharmacological stressor yohimbine did not reinstate suboptimal choice, but did increase impulsive action as indexed by premature responding. Presentation of cues previously associated with large wins did not alter behavior. Results suggest reinstaters of suboptimal choice may differ from reinstaters of extinguished drug- and food-seeking behavior.

12.
Exp Clin Psychopharmacol ; 22(6): 557-64, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25330246

RESUMO

The Attentional Set-Shifting Task (ASST) is a rodent analog of the Wisconsin Card Sorting Task, which measures executive functioning. The ASST tests for reversal of stimulus-response learning and the formation and maintenance of attentional sets. Depletion of dopamine has been shown to improve performance on attentional shifts. The study presented here questioned whether a D4-specific antagonist, L-745,870, could have a similar effect on animals, even after being treated with repeated doses of amphetamine. Three groups of male rats were given either 10 saline injections (n = 12), 10 amphetamine injections (2 mg/kg; n = 8), or 10 amphetamine injections plus 1 pretreatment injection of L-745,870 (0.1 mg/kg; n = 8) 20 min prior to testing. One-way ANOVA results showed that amphetamine-only rats were impaired on all 3 reversals (Ms = 19, 16.4, and 17.1) compared with L-745,870-treated rats (Ms = 9.8, 10.9, and 9.6) and controls (Ms = 8.6, 9.6, 9.3; all ps < .01). L-745,870-treated rats also displayed reduced latencies to respond compared with both saline controls and amphetamine rats. It is thought that D4 receptors play a role in cue salience, and that by blocking these receptors, animals display less attachment to previously rewarded cues. The results presented support this idea and imply that blocking of D4 receptors can reverse the impairment in reversals caused by amphetamine.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/tratamento farmacológico , Anfetamina/toxicidade , Antagonistas de Dopamina/uso terapêutico , Deficiências da Aprendizagem/prevenção & controle , Nootrópicos/uso terapêutico , Receptores de Dopamina D4/antagonistas & inibidores , Reversão de Aprendizagem/efeitos dos fármacos , Anfetamina/administração & dosagem , Transtornos Relacionados ao Uso de Anfetaminas/fisiopatologia , Animais , Atenção/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/administração & dosagem , Estimulantes do Sistema Nervoso Central/toxicidade , Sinais (Psicologia) , Relação Dose-Resposta a Droga , Função Executiva/efeitos dos fármacos , Deficiências da Aprendizagem/etiologia , Masculino , Terapia de Alvo Molecular , Piridinas/uso terapêutico , Pirróis/uso terapêutico , Ratos Long-Evans , Recompensa , Índice de Gravidade de Doença
13.
Behav Brain Res ; 210(1): 123-6, 2010 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-20122971

RESUMO

Intermittent hypoxia (IH), a characteristic of sleep apnea, was modeled in Fischer Brown Norway rats (10h/day for 7 days) followed by cognitive testing in an attentional set-shifting task. The ability to shift attention from one sensory modality (e.g., odor) to another (e.g., digging medium) was impaired, a finding that could not be attributed to deficits in attention, discrimination, learning, or motor performance. Instead, the deficit is likely to reflect impaired allocation of attentional resources of the working memory system.


Assuntos
Atenção , Transtornos Cognitivos/etiologia , Função Executiva , Hipóxia/complicações , Animais , Cognição , Discriminação Psicológica , Modelos Animais de Doenças , Aprendizagem , Masculino , Memória de Curto Prazo , Atividade Motora , Testes Neuropsicológicos , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Síndromes da Apneia do Sono , Fatores de Tempo
14.
Brain Res ; 1294: 128-37, 2009 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-19643093

RESUMO

Obstructive sleep apnea is primarily characterized by hypoxemia due to frequent apneic episodes and fragmentation of sleep due to the brief arousals that terminate the apneic episodes. Though neurobehavioral deficits frequently accompany sleep apnea, the relative roles of hypoxia versus sleep fragmentation are difficult to separate in apneic patients. Here, we assessed cognitive function as measured by water maze in the Fischer/Brown Norway (FBN) rat, comparing 24 h of sleep interruption (SI) to 24 h of intermittent hypoxia (IH), in order to dissociate their relative contributions to cognitive impairment. For SI, automated treadmills were used to induce brief ambulation in rats every 2 min, either prior to, or after, initial water maze acquisition training. IH was simulated by cycling environmental oxygen levels between 6% and 19% every 2 min, again either prior to, or after, acquisition. Twenty-four hours of IH exposure had no significant effect on either acquisition or retention, irrespective of whether IH occurred prior to, or after, acquisition. To replicate previous work, another group of rats, exposed to 3 days of IH (10 h/day) prior to acquisition, had impaired performance during acquisition. A comparison of the 24 h IH and 3 day IH findings suggests that a minimum amount of IH exposure is necessary to produce detectable spatial memory impairments. Although SI before acquisition had no effect on acquisition or later retention of the hidden platform location, SI after acquisition robustly impaired retention, indicating that spatial memory consolidation is more susceptible to the effects of sleep disruption than is the acquisition (learning) of spatial information.


Assuntos
Hipóxia/fisiopatologia , Deficiências da Aprendizagem/fisiopatologia , Transtornos da Memória/fisiopatologia , Apneia Obstrutiva do Sono/fisiopatologia , Privação do Sono/fisiopatologia , Percepção Espacial/fisiologia , Animais , Modelos Animais de Doenças , Hipóxia/complicações , Deficiências da Aprendizagem/etiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/etiologia , Oxigênio/metabolismo , Ratos , Ratos Endogâmicos F344 , Apneia Obstrutiva do Sono/complicações , Privação do Sono/complicações , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa