Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Radiat Res ; 183(3): 367-74, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25688996

RESUMO

Fractionated whole-brain irradiation (fWBI), used to treat brain metastases, often leads to neurologic injury and cognitive impairment. The cognitive effects of irradiation in nonhuman primates (NHP) have been previously published; this report focuses on corresponding neuropathologic changes that could have served as the basis for those effects in the same study. Four rhesus monkeys were exposed to 40 Gy of fWBI [5 Gy × 8 fraction (fx), 2 fx/week for four weeks] and received anatomical MRI prior to, and 14 months after fWBI. Neurologic and histologic sequelae were studied posthumously. Three of the NHPs underwent cognitive assessments, and each exhibited radiation-induced impairment associated with various degrees of vascular and inflammatory neuropathology. Two NHPs had severe multifocal necrosis of the forebrain, midbrain and brainstem. Histologic and MRI findings were in agreement, and the severity of cognitive decrement previously reported corresponded to the degree of observed pathology in two of the animals. In response to fWBI, the NHPs showed pathology similar to humans exposed to radiation and show comparable cognitive decline. These results provide a basis for implementing NHPs to examine and treat adverse cognitive and neurophysiologic sequelae of radiation exposure in humans.


Assuntos
Neoplasias Encefálicas/radioterapia , Encéfalo/patologia , Transtornos Cognitivos/fisiopatologia , Macaca mulatta , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos da radiação , Neoplasias Encefálicas/patologia , Transtornos Cognitivos/diagnóstico por imagem , Fracionamento da Dose de Radiação , Humanos , Radiografia , Irradiação Corporal Total
2.
Radiat Res ; 182(5): 580-5, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25338095

RESUMO

Future space missions are expected to include increased extravehicular activities (EVAs) during which astronauts are exposed to high-energy space radiation while breathing 100% oxygen. Given that brain irradiation can lead to cognitive impairment, and that oxygen is a potent radiosensitizer, there is a concern that astronauts may be at greater risk of developing cognitive impairment when exposed to space radiation while breathing 100% O(2) during an EVA. To address this concern, unanesthetized, unrestrained, young adult male Fischer 344 × Brown Norway rats were allowed to breathe 100% O(2) for 30 min prior to, during and 2 h after whole-body irradiation with 0, 1, 3, 5 or 7 Gy doses of 18 MV X rays delivered from a medical linear accelerator at a dose rate of ~425 mGy/min. Irradiated and unirradiated rats breathing air (~21% O(2)) served as controls. Cognitive function was assessed 9 months postirradiation using the perirhinal cortex-dependent novel object recognition task. Cognitive function was not impaired until the rats breathing either air or 100% O(2) received a whole-body dose of 7 Gy. However, at all doses, cognitive function of the irradiated rats breathing 100% O(2) was improved over that of the irradiated rats breathing air. These data suggest that astronauts are not at greater risk of developing cognitive impairment when exposed to space radiation while breathing 100% O(2) during an EVA.


Assuntos
Transtornos Cognitivos/etiologia , Radiação Cósmica/efeitos adversos , Oxigênio/administração & dosagem , Animais , Masculino , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344 , Respiração , Voo Espacial
3.
Int J Radiat Biol ; 90(9): 799-806, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24991879

RESUMO

PURPOSE: To assess the long-term effects of fractionated whole brain irradiation (fWBI) using diffusion tensor imaging (DTI) and behavior in a pediatric rodent model for the clinical presentation of adult pediatric cancer survivors. MATERIALS AND METHODS: Five-week-old, male F344xBN rats were randomized to receive 0, 5, or 6.5 Gy fractions biweekly for 3 weeks, resulting in Sham, Irradiated-30 (IR-30) and IR-39 Gy total dose groups. Magnetic Resonance Imaging occurred at 1, 3, 6 and 9 months with behavioral assessment at 10-11 months post-fWBI. RESULTS: Irradiation reduced brain size (p < 0.001) and body weight (p < 0.001) proportionate to dose. At 1 month post-fWBI and throughout follow-up, diffusion was reduced in IR-30 and IR-39 relative to shams (p < 0.001). IR-30 but not IR-39 rats were impaired relative to Shams on the reversal trial of the Morris Water Maze (p < 0.05), and IR-30 rats preferred a striatum- mediated strategy (p < 0.06). CONCLUSIONS: Hippocampal performance was impaired in IR-30 but not IR-39 animals. While gross size differences exist, white matter integrity is preserved in rats after fWBI at 5 weeks. This significant departure from childhood cancer survivors and single fraction rodent studies where white matter degradation is a prominent feature are discussed.


Assuntos
Encéfalo/efeitos da radiação , Cognição/efeitos da radiação , Imagem de Tensor de Difusão , Fracionamento da Dose de Radiação , Radioterapia/métodos , Animais , Comportamento Animal/efeitos da radiação , Neoplasias Encefálicas/radioterapia , Hipocampo/efeitos da radiação , Imageamento por Ressonância Magnética , Masculino , Aprendizagem em Labirinto , Modelos Animais , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344
4.
Int J Radiat Biol ; 90(9): 790-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24937374

RESUMO

PURPOSE: To determine if the brain's response to single doses predicts its response to 'biologically equivalent' fractionated doses. METHODS: Young adult male Fischer 344 rats were whole-brain irradiated with either single 11, 14, or 16.5 Gy doses of (137)Cs γ rays or their 'biologically equivalent' 20, 30, or 40 Gy fractionated doses (fWBI) delivered in 5 Gy fractions, twice/week for 2, 3, or 4 weeks, respectively. At 2 months post-irradiation, cellular markers of inflammation (total, activated, and newborn microglia) and neurogenesis (newborn neurons) were measured in 40 µm sections of the dentate gyrus (DG). RESULTS: Although the total number of microglia in the DG/hilus was not significantly different (p > 0.7) in unirradiated, single dose, and fWBI rats, single doses produced a significant (p < 0.003) increase in the percent-activated microglia; fWBI did not (p > 0.1). Additionally, single doses produced a significant (p < 0.002) dose-dependent increase in surviving newborn microglia; fWBI did not (p < 0.8). Although total proliferation in the DG was reduced equally by single and fWBI doses, single doses produced a significant dose-dependent (p < 0.02) decrease in surviving newborn neurons; fWBI did not (p > 0.6). CONCLUSIONS: These data demonstrate that the rat brain's cellular response to single doses often does not predict its cellular response to 'biologically equivalent' fWBI doses.


Assuntos
Encéfalo/diagnóstico por imagem , Radioisótopos de Césio/química , Animais , Proliferação de Células/efeitos da radiação , Giro Denteado/efeitos da radiação , Fracionamento da Dose de Radiação , Relação Dose-Resposta à Radiação , Raios gama , Hipocampo/efeitos da radiação , Inflamação/radioterapia , Masculino , Microglia/patologia , Neurogênese/efeitos da radiação , Neurônios/efeitos da radiação , Cintilografia , Ratos , Ratos Endogâmicos F344
5.
Radiat Res ; 181(1): 33-44, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24397438

RESUMO

We hypothesized that dietary administration of the peroxisomal proliferator-activated receptor α agonist, fenofibrate, to young adult male rats would prevent the fractionated whole-brain irradiation (fWBI)-induced reduction in cognitive function and neurogenesis and prevent the fWBI-induced increase in the total number of activated microglia. Eighty 12-14-week-old young adult male Fischer 344 × Brown Norway rats received either: (1) sham irradiation, (2) 40 Gy of fWBI delivered as two 5 Gy fractions/week for 4 weeks, (3) sham irradiation + dietary fenofibrate (0.2% w/w) starting 7 days prior to irradiation, or (4) fWBI + fenofibrate. Cognitive function was measured 26-29 weeks after irradiation using: (1) the perirhinal cortex (PRh)-dependent novel object recognition task; (2) the hippocampal-dependent standard Morris water maze (MWM) task; (3) the hippocampal-dependent delayed match-to-place version of the MWM task; and (4) a cue strategy preference version of the MWM to distinguish hippocampal from striatal task performance. Neurogenesis was assessed 29 weeks after fWBI in the granular cell layer and subgranular zone of the dentate gyrus using a doublecortin antibody. Microglial activation was assessed using an ED1 antibody in the dentate gyrus and hilus of the hippocampus. A significant impairment in perirhinal cortex-dependent cognitive function was measured after fWBI. In contrast, fWBI failed to alter hippocampal-dependent cognitive function, despite a significant reduction in hippocampal neurogenesis. Continuous administration of fenofibrate prevented the fWBI-induced reduction in perirhinal cortex-dependent cognitive function, but did not prevent the radiation-induced reduction in neurogenesis or the radiation-induced increase in activated microglia. These data suggest that fenofibrate may be a promising therapeutic for the prevention of some modalities of radiation-induced cognitive impairment in brain cancer patients.


Assuntos
Cognição/efeitos dos fármacos , Cognição/efeitos da radiação , Fenofibrato/farmacologia , PPAR alfa/agonistas , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/efeitos da radiação , Peso Corporal/efeitos dos fármacos , Peso Corporal/efeitos da radiação , Proteína Duplacortina , Masculino , Atividade Motora/efeitos dos fármacos , Atividade Motora/efeitos da radiação , Ratos , Acuidade Visual/efeitos dos fármacos , Acuidade Visual/efeitos da radiação
6.
Radiat Res ; 181(1): 21-32, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24377717

RESUMO

Fractionated partial or whole-brain irradiation is the primary treatment for metastatic brain tumors. Despite reducing tumor burden and increasing lifespan, progressive, irreversible cognitive impairment occurs in >50% of the patients who survive >6 months after fractionated whole-brain irradiation. The exact mechanism(s) responsible for this radiation-induced brain injury are unknown; however, preclinical studies suggest that radiation modulates the extracellular receptor kinase signaling pathway, which is associated with cognitive impairment in many neurological diseases. In the study reported here, we demonstrated that the extracellular receptor kinase transcriptionally-regulated early response gene, Homer1a, was up-regulated transiently in the hippocampus and down-regulated in the cortex of young adult male Fischer 344 X Brown Norway rats at 48 h after 40 Gy of fractionated whole-brain irradiation. Two months after fractionated whole-brain irradiation, these changes in Homer1a expression correlated with a down-regulation of the hippocampal glutamate receptor 1 and protein kinase Cγ, and an up-regulation of cortical glutamate receptor 1 and protein kinase Cγ. Two drugs that prevent radiation-induced cognitive impairment in rats, the angiotensin type-1 receptor blocker, L-158,809, and the angiotensin converting enzyme inhibitor, ramipril, reversed the fractionated whole-brain irradiation-induced Homer1a expression at 48 h in the hippocampus and cortex and restored glutamate receptor 1 and protein kinase Cγ to the levels in sham-irradiated controls at 2 months after fractionated whole-brain irradiation. These data indicate that Homer1a is, (1) a brain region specific regulator of radiation-induced brain injury, including cognitive impairment and (2) potentially a druggable target for preventing it.


Assuntos
Lesões Encefálicas/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/efeitos da radiação , Hipocampo/metabolismo , Hipocampo/efeitos da radiação , Lesões por Radiação/metabolismo , Animais , Lesões Encefálicas/genética , Cognição/efeitos dos fármacos , Cognição/efeitos da radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Proteínas de Arcabouço Homer , Imidazóis/farmacologia , Masculino , Proteína Quinase C/metabolismo , Lesões por Radiação/genética , Ramipril/farmacologia , Ratos , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Tetrazóis/farmacologia , Fatores de Tempo
7.
Redox Biol ; 1: 70-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24024139

RESUMO

Glioma survival is dismal, in part, due to an imbalance in antioxidant expression and activity. Peroxisome proliferator-activated receptor (PPAR) agonists have antineoplastic properties which present new redox-dependent targets for glioma anticancer therapies. Herein, we demonstrate that treatment of primary cultures of normal rat astrocytes with PPAR agonists increased the expression of catalase mRNA protein, and enzymatic activity. In contrast, these same agonists had no effect on catalase expression and activity in malignant rat glioma cells. The increase in steady-state catalase mRNA observed in normal rat astrocytes was due, in part, to de novo mRNA synthesis as opposed to increased catalase mRNA stability. Moreover, pioglitazone-mediated induction of catalase activity in normal rat astrocytes was completely blocked by transfection with a PPARγ-dominant negative plasmid. These data suggest that defects in PPAR-mediated signaling and gene expression may represent a block to normal catalase expression and induction in malignant glioma. The ability of PPAR agonists to differentially increase catalase expression and activity in normal astrocytes but not glioma cells suggests that these compounds might represent novel adjuvant therapeutic agents for the treatment of gliomas.


Assuntos
Astrócitos/efeitos dos fármacos , Catalase/genética , Catalase/metabolismo , Glioma/genética , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Células COS , Chlorocebus aethiops , Regulação da Expressão Gênica/efeitos dos fármacos , Glioma/metabolismo , Humanos , Pioglitazona , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Células Tumorais Cultivadas
8.
Free Radic Biol Med ; 65: 1060-1068, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24012919

RESUMO

About 500,000 new cancer patients will develop brain metastases in 2013. The primary treatment modality for these patients is partial or whole brain irradiation which leads to a progressive, irreversible cognitive impairment. Although the exact mechanisms behind this radiation-induced brain injury are unknown, neuroinflammation in glial populations is hypothesized to play a role. Blockers of the renin-angiotensin system (RAS) prevent radiation-induced cognitive impairment and modulate radiation-induced neuroinflammation. Recent studies suggest that RAS blockers may reduce inflammation by increasing endogenous concentrations of the anti-inflammatory heptapeptide angiotensin-(1-7) [Ang-(1-7)]. Ang-(1-7) binds to the AT(1-7) receptor and inhibits MAP kinase activity to prevent inflammation. This study describes the inflammatory response to radiation in astrocytes characterized by radiation-induced increases in (i) IL-1ß and IL-6 gene expression; (ii) COX-2 and GFAP immunoreactivity; (iii) activation of AP-1 and NF-κB transcription factors; and (iv) PKCα, MEK, and ERK (MAP kinase) activation. Treatment with U-0126, a MEK inhibitor, demonstrates that this radiation-induced inflammation in astrocytes is mediated through the MAP kinase pathway. Ang-(1-7) inhibits radiation-induced inflammation, increases in PKCα, and MAP kinase pathway activation (phosphorylation of MEK and ERK). Additionally Ang-(1-7) treatment leads to an increase in dual specificity phosphatase 1 (DUSP1). Furthermore, treatment with sodium vanadate (Na3VO4), a phosphatase inhibitor, blocks Ang-(1-7) inhibition of radiation-induced inflammation and MAP kinase activation, suggesting that Ang-(1-7) alters phosphatase activity to inhibit radiation-induced inflammation. These data suggest that RAS blockers inhibit radiation-induced inflammation and prevent radiation-induced cognitive impairment not only by reducing Ang II but also by increasing Ang-(1-7) levels.


Assuntos
Angiotensina I/farmacologia , Astrócitos/imunologia , Sistema de Sinalização das MAP Quinases , Fragmentos de Peptídeos/farmacologia , Protetores contra Radiação/farmacologia , Animais , Astrócitos/efeitos da radiação , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Fosfatase 1 de Especificidade Dupla/metabolismo , Inflamação/metabolismo , Cultura Primária de Células , Ratos
9.
Free Radic Biol Med ; 61: 1-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23499837

RESUMO

Brain tumor patients often develop cognitive impairment months to years after partial or fractionated whole-brain irradiation (WBI). Studies suggest that neuroinflammation and decreased hippocampal neurogenesis contribute to the pathogenesis of radiation-induced brain injury. In this study, we determined if the peroxisomal proliferator-activated receptor (PPAR) δ agonist GW0742 can prevent radiation-induced brain injury in C57Bl/6 wild-type (WT) and PPARδ knockout (KO) mice. Dietary GW0742 prevented the acute increase in IL-1ß mRNA and ERK phosphorylation measured at 3h after a single 10-Gy dose of WBI; it also prevented the increase in the number of activated hippocampal microglia 1 week after WBI. In contrast, dietary GW074 failed to prevent the radiation-induced decrease in hippocampal neurogenesis determined 2 months after WBI in WT mice or to mitigate their hippocampal-dependent spatial memory impairment measured 3 months after WBI using the Barnes maze task. PPARδ KO mice exhibited defects including decreased numbers of astrocytes in the dentate gyrus/hilus of the hippocampus and a failure to exhibit a radiation-induced increase in activated hippocampal microglia. Interestingly, the number of astrocytes in the dentate gyrus/hilus was reduced in WT mice, but not in PPARδ KO mice 2 months after WBI. These results demonstrate that, although dietary GW0742 prevents the increase in inflammatory markers and hippocampal microglial activation in WT mice after WBI, it does not restore hippocampal neurogenesis or prevent early delayed hippocampal-dependent cognitive impairment after WBI. Thus, the exact relationship between radiation-induced neuroinflammation, neurogenesis, and cognitive impairment remains elusive.


Assuntos
Transtornos Cognitivos/prevenção & controle , Irradiação Craniana/efeitos adversos , Hipocampo/efeitos da radiação , Neurogênese/efeitos dos fármacos , PPAR delta/agonistas , Tiazóis/farmacologia , Animais , Transtornos Cognitivos/etiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipocampo/patologia , Hipocampo/fisiologia , Inflamação/prevenção & controle , Interleucina-1beta/genética , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/efeitos da radiação , Fosforilação
10.
Neurology ; 80(8): 747-53, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23390169

RESUMO

OBJECTIVE: In a retrospective review to assess neuroanatomical targets of radiation-induced cognitive decline, dose volume histogram (DVH) analyses of specific brain regions of interest (ROI) are correlated to neurocognitive performance in 57 primary brain tumor survivors. METHODS: Neurocognitive assessment at baseline included Trail Making Tests A/B, a modified Rey-Osterreith Complex Figure, California or Hopkins Verbal Learning Test, Digit Span, and Controlled Oral Word Association. DVH analysis was performed for multiple neuroanatomical targets considered to be involved in cognition. The %v10 (percent of ROI receiving 10 Gy), %v40, and %v60 were calculated for each ROI. Factor analysis was used to estimate global cognition based on a summary of performance on individual cognitive tests. Stepwise regression was used to determine which dose volume predicted performance on global factors and individual neurocognitive tests for each ROI. RESULTS: Regions that predicted global cognitive outcomes at doses <60 Gy included the corpus callosum, left frontal white matter, right temporal lobe, bilateral hippocampi, subventricular zone, and cerebellum. Regions of adult neurogenesis primarily predicted cognition at %v40 except for the right hippocampus which predicted at %v10. Regions that did not predict global cognitive outcomes at any dose include total brain volume, frontal pole, anterior cingulate, right frontal white matter, and the right precentral gyrus. CONCLUSIONS: Modeling of radiation-induced cognitive decline using neuroanatomical target theory appears to be feasible. A prospective trial is necessary to validate these data.


Assuntos
Encéfalo/efeitos da radiação , Transtornos Cognitivos/etiologia , Lesões por Radiação/complicações , Radioterapia/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Transtornos Cognitivos/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Lesões por Radiação/patologia , Análise de Regressão , Sobreviventes
11.
Clin Cancer Res ; 19(9): 2294-300, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23388505

RESUMO

Each year, approximately 200,000 patients in the United States will receive partial- or whole-brain irradiation for the treatment of primary or metastatic brain cancer. Early and delayed radiation effects are transient and reversible with modern therapeutic standards; yet, late radiation effects (≥6 months postirradiation) remain a significant risk, resulting in progressive cognitive impairment. These risks include functional deficits in memory, attention, and executive function that severely affect the patient's quality of life. The mechanisms underlying radiation-induced cognitive impairment remain ill defined. Classically, radiation-induced alterations in vascular and neuroinflammatory glial cell clonogenic populations were hypothesized to be responsible for radiation-induced brain injury. Recently, preclinical studies have focused on the hippocampus, one of two sites of adult neurogenesis within the brain, which plays an important role in learning and memory. Radiation ablates hippocampal neurogenesis, alters neuronal function, and induces neuroinflammation. Neuronal stem cells implanted into the hippocampus prevent the decrease in neurogenesis and improve cognition after irradiation. Clinically prescribed drugs, including PPARα and PPARγ agonists, as well as RAS blockers, prevent radiation-induced neuroinflammation and cognitive impairment independent of improved neurogenesis. Translating these exciting findings to the clinic offers the promise of improving the quality of life of brain tumor patients who receive radiotherapy.


Assuntos
Transtornos Cognitivos/patologia , Lesões por Radiação/patologia , Animais , Encéfalo/patologia , Encéfalo/efeitos da radiação , Neoplasias Encefálicas/radioterapia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/prevenção & controle , Fenofibrato/farmacologia , Humanos , Neurogênese/efeitos da radiação , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Lesões por Radiação/etiologia , Lesões por Radiação/prevenção & controle , Lesões Experimentais por Radiação/patologia , Protetores contra Radiação/farmacologia , Sistema Renina-Angiotensina
12.
Methods Mol Biol ; 952: 325-36, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23100244

RESUMO

The use of behavioral testing has become an invaluable tool for assessing the efficacy of therapeutics for a variety of disorders of the central nervous system. This chapter will describe in detail several behavioral paradigms to evaluate the efficacy of PPAR agonists to modulate cognitive impairments in rodent models. When used together as a battery these procedures allow for a global assessment of cognition. These tests are explained in detail below, and include: (1) Novel Object Recognition (NOR), (2) Morris Water Maze (MWM), (3) Delay Match to Place (DMP), and (4) Cue Strategy.


Assuntos
Comportamento Animal/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Testes Neuropsicológicos , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Animais , Lesões Encefálicas/fisiopatologia , Sinais (Psicologia) , Modelos Animais de Doenças , Aprendizagem em Labirinto/efeitos dos fármacos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Resultado do Tratamento
13.
Neuro Oncol ; 14 Suppl 4: iv37-44, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23095829

RESUMO

Approximately 100,000 patients per year in the United States with primary and metastatic brain tumor survive long enough (>6 months) to develop radiation-induced brain injury. Before 1970, the human brain was thought to be radioresistant; the acute central nervous system (CNS) syndrome occurs after single doses of ≥ 30 Gy, and white matter necrosis can occur at fractionated doses of ≥ 60 Gy. Although white matter necrosis is uncommon with modern radiation therapy techniques, functional deficits, including progressive impairments in memory, attention, and executive function have become increasingly important, having profound effects on quality of life. Preclinical studies have provided valuable insights into the pathogenic mechanisms involved in radiation-induced cognitive impairment. Although reductions in hippocampal neurogenesis and hippocampal-dependent cognitive function have been observed in rodent models, it is important to recognize that other brain regions are affected; non-hippocampal-dependent reductions in cognitive function occur. Neuroinflammation is viewed as playing a major role in radiation-induced cognitive impairment. During the past 5 years, several preclinical studies have demonstrated that interventional therapies aimed at modulating neuroinflammation can prevent/ameliorate radiation-induced cognitive impairment independent of changes in neurogenesis. Translating these exciting preclinical findings to the clinic offers the promise of improving the quality of life in patients with brain tumors who receive radiation therapy.


Assuntos
Neoplasias Encefálicas/radioterapia , Encéfalo/efeitos da radiação , Transtornos Cognitivos/etiologia , Lesões por Radiação/complicações , Radioterapia/efeitos adversos , Animais , Humanos
14.
Front Oncol ; 2: 73, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22833841

RESUMO

Approximately 100,000 primary and metastatic brain tumor patients/year in the US survive long enough (>6 months) to experience radiation-induced brain injury. Prior to 1970, the human brain was thought to be highly radioresistant; the acute CNS syndrome occurs after single doses >30 Gy; white matter necrosis occurs at fractionated doses >60 Gy. Although white matter necrosis is uncommon with modern techniques, functional deficits, including progressive impairments in memory, attention, and executive function have become important, because they have profound effects on quality of life. Preclinical studies have provided valuable insights into the pathogenesis of radiation-induced cognitive impairment. Given its central role in memory and neurogenesis, the majority of these studies have focused on the hippocampus. Irradiating pediatric and young adult rodent brains leads to several hippocampal changes including neuroinflammation and a marked reduction in neurogenesis. These data have been interpreted to suggest that shielding the hippocampus will prevent clinical radiation-induced cognitive impairment. However, this interpretation may be overly simplistic. Studies using older rodents, that more closely match the adult human brain tumor population, indicate that, unlike pediatric and young adult rats, older rats fail to show a radiation-induced decrease in neurogenesis or a loss of mature neurons. Nevertheless, older rats still exhibit cognitive impairment. This occurs in the absence of demyelination and/or white matter necrosis similar to what is observed clinically, suggesting that more subtle molecular, cellular and/or microanatomic modifications are involved in this radiation-induced brain injury. Given that radiation-induced cognitive impairment likely reflects damage to both hippocampal- and non-hippocampal-dependent domains, there is a critical need to investigate the microanatomic and functional effects of radiation in various brain regions as well as their integration at clinically relevant doses and schedules. Recently developed techniques in neuroscience and neuroimaging provide not only an opportunity to accomplish this, but they also offer the opportunity to identify new biomarkers and new targets for interventions to prevent or ameliorate these late effects.

15.
Radiat Res ; 178(1): 46-56, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22687052

RESUMO

We hypothesized that chronic administration of the angiotensin-converting enzyme inhibitor, ramipril, to young adult male rats would prevent/ameliorate fractionated whole-brain irradiation-induced perirhinal cortex-dependent cognitive impairment. Eighty 12-14-week-old young adult male Fischer 344 rats received either: (1) sham irradiation, (2) 40 Gy of fractionated whole-brain irradiation delivered as two 5 Gy fractions/week for 4 weeks, (3) sham irradiation plus continuous administration of 15 mg/L of ramipril in the drinking water starting 3 days before irradiation, or (4) fractionated whole-brain irradiation plus ramipril. Cognitive function was assessed using a perirhinal cortex-dependent version of the novel object recognition task 26 weeks after irradiation. Microglial activation was determined in the perirhinal cortex and the dentate gyrus of the hippocampus 28 weeks after irradiation using the ED1 antibody. Neurogenesis was assessed in the granular cell layer and subgranular zones of the dentate gyrus using a doublecortin antibody. Fractionated whole-brain irradiation led to: (1) a significant impairment in perirhinal cortex-dependent cognitive function, (2) a significant increase in activated microglia in the dentate gyrus but not in the perirhinal cortex, and (3) a significant decrease in neurogenesis. Continuous administration of ramipril before, during, and after irradiation prevented the fractionated whole-brain irradiation-induced changes in perirhinal cortex-dependent cognitive function, as well as in microglial activation in the dentate gyrus. Thus, as hypothesized, continuous administration of the angiotensin-converting enzyme inhibitor, ramipril, can prevent the fractionated whole-brain irradiation-induced impairment in perirhinal cortex-dependent cognitive function.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Córtex Cerebral/efeitos da radiação , Transtornos Cognitivos/prevenção & controle , Irradiação Craniana/efeitos adversos , Lesões Experimentais por Radiação/prevenção & controle , Ramipril/uso terapêutico , Angiotensina I/sangue , Animais , Peso Corporal , Córtex Cerebral/fisiologia , Transtornos Cognitivos/etiologia , Fracionamento da Dose de Radiação , Proteína Duplacortina , Masculino , Ratos , Ratos Endogâmicos F344
16.
Free Radic Biol Med ; 52(9): 1734-43, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22387176

RESUMO

Partial or whole-brain irradiation is often required to treat both primary and metastatic brain cancer. Radiation-induced normal tissue injury, including progressive cognitive impairment, however, can significantly affect the well-being of the approximately 200,000 patients who receive these treatments each year in the United States. Although the exact mechanisms underlying radiation-induced late effects remain unclear, oxidative stress and inflammation are thought to play a critical role. Microglia are key mediators of neuroinflammation. Peroxisomal proliferator-activated receptor (PPAR) δ has been shown to be a potent regulator of anti-inflammatory responses. Thus, we hypothesized that PPARδ activation would modulate the radiation-induced inflammatory response in microglia. Incubating BV-2 murine microglial cells with the PPARδ agonist L-165041 prevented the radiation-induced increase in: (i) intracellular reactive oxygen species generation, (ii) Cox-2 and MCP-1 expression, and (iii) IL-1ß and TNF-α message levels. This occurred, in part, through PPARδ-mediated modulation of stress-activated kinases and proinflammatory transcription factors. PPARδ inhibited NF-κB via transrepression by physically interacting with the p65 subunit and prevented activation of the PKCα/MEK1/2/ERK1/2/AP-1 pathway by inhibiting the radiation-induced increase in intracellular reactive oxygen species generation. These data support the hypothesis that PPARδ activation can modulate radiation-induced oxidative stress and inflammatory responses in microglia.


Assuntos
Microglia/patologia , NF-kappa B/antagonistas & inibidores , PPAR delta/fisiologia , Proteínas Quinases/metabolismo , Fator de Transcrição AP-1/antagonistas & inibidores , Animais , Sequência de Bases , Western Blotting , Linhagem Celular Transformada , Ensaio de Desvio de Mobilidade Eletroforética , Imunoprecipitação , Inflamação/etiologia , Inflamação/prevenção & controle , Camundongos , Microglia/metabolismo , Reação em Cadeia da Polimerase , RNA/genética , Espécies Reativas de Oxigênio/metabolismo
17.
Radiat Res ; 177(4): 449-66, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22348250

RESUMO

Technological developments in radiation therapy and other cancer therapies have led to a progressive increase in five-year survival rates over the last few decades. Although acute effects have been largely minimized by both technical advances and medical interventions, late effects remain a concern. Indeed, the need to identify those individuals who will develop radiation-induced late effects, and to develop interventions to prevent or ameliorate these late effects is a critical area of radiobiology research. In the last two decades, preclinical studies have clearly established that late radiation injury can be prevented/ameliorated by pharmacological therapies aimed at modulating the cascade of events leading to the clinical expression of radiation-induced late effects. These insights have been accompanied by significant technological advances in imaging that are moving radiation oncology and normal tissue radiobiology from disciplines driven by anatomy and macrostructure to ones in which important quantitative functional, microstructural, and metabolic data can be noninvasively and serially determined. In the current article, we review use of positron emission tomography (PET), single photon emission tomography (SPECT), magnetic resonance (MR) imaging and MR spectroscopy to generate pathophysiological and functional data in the central nervous system, lung, and heart that offer the promise of, (1) identifying individuals who are at risk of developing radiation-induced late effects, and (2) monitoring the efficacy of interventions to prevent/ameliorate them.


Assuntos
Diagnóstico por Imagem/métodos , Lesões por Radiação/diagnóstico , Animais , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/etiologia , Irradiação Craniana/efeitos adversos , Imagem de Tensor de Difusão/métodos , Traumatismos Cardíacos/diagnóstico , Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/patologia , Humanos , Imageamento por Ressonância Magnética/métodos , Espectroscopia de Ressonância Magnética/métodos , Neuroimagem/métodos , Imagem de Perfusão/métodos , Tomografia por Emissão de Pósitrons/métodos , Medicina de Precisão , Lesões por Radiação/diagnóstico por imagem , Lesões por Radiação/patologia , Lesões Experimentais por Radiação/diagnóstico , Lesões Experimentais por Radiação/diagnóstico por imagem , Lesões Experimentais por Radiação/patologia , Pneumonite por Radiação/diagnóstico , Pneumonite por Radiação/diagnóstico por imagem , Pneumonite por Radiação/etiologia , Tolerância a Radiação , Radiografia , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Doenças Vasculares/etiologia , Doenças Vasculares/patologia
18.
PPAR Res ; 2011: 373560, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22135673

RESUMO

Peroxisome proliferator-activated receptors (PPARα, δ, and γ) are ligand-activated transcription factors that regulate a wide range of cellular processes, including inflammation, proliferation, differentiation, metabolism, and energy homeostasis. All three PPAR subtypes have been identified in the central nervous system (CNS) of rodents. While PPARα and PPARγ are expressed in more restricted areas of the CNS, PPARδ is ubiquitously expressed and is the predominant subtype. Although data regarding PPARδ are limited, studies have demonstrated that administration of PPARδ agonists confers neuroprotection following various acute and chronic injuries to the CNS, such as stroke, multiple sclerosis, and Alzheimer's disease. The antioxidant and anti-inflammatory properties of PPARδ agonists are thought to underly their neuroprotective efficacy. This review will focus on the putative neuroprotective benefits of therapeutically targeting PPARδ in the CNS, and specifically, highlight the antioxidant and anti-inflammatory functions of PPARδ agonists.

19.
Int J Radiat Biol ; 87(10): 1001-10, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21961967

RESUMO

PURPOSE: In response to the increased risk of radiological terrorist attack, a network of Centers for Medical Countermeasures against Radiation (CMCR) has been established in the United States, focusing on evaluating animal model responses to uniform, relatively homogenous whole- or partial-body radiation exposures at relatively high dose rates. The success of such studies is dependent not only on robust animal models but on accurate and reproducible dosimetry within and across CMCR. To address this issue, the Education and Training Core of the Duke University School of Medicine CMCR organised a one-day workshop on small animal dosimetry. Topics included accuracy in animal dosimetry accuracy, characteristics and differences of cesium-137 and X-ray irradiators, methods for dose measurement, and design of experimental irradiation geometries for uniform dose distributions. This paper summarises the information presented and discussed. CONCLUSIONS: Without ensuring accurate and reproducible dosimetry the development and assessment of the efficacy of putative countermeasures will not prove successful. Radiation physics support is needed, but is often the weakest link in the small animal dosimetry chain. We recommend: (i) A user training program for new irradiator users, (ii) subsequent training updates, and (iii) the establishment of a national small animal dosimetry center for all CMCR members.


Assuntos
Modelos Animais , Radiometria/métodos , Animais , Calibragem , Radioisótopos de Césio , Radiometria/instrumentação , Radiometria/normas , Reprodutibilidade dos Testes , Medição de Risco , Sensibilidade e Especificidade , Raios X
20.
Radiat Res ; 175(4): 519-25, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21275607

RESUMO

To investigate the effect of fractionated whole-brain irradiation on nonhuman primates, 6-9-year-old male rhesus monkeys were irradiated with 40 Gy delivered as two 5-Gy fractions/week for 4 weeks. Cognitive function was assessed 5 days/week for 4 months prior to fractionated whole-brain irradiation and for 11 months after irradiation using a Delayed-Match-to-Sample (DMS) task at both low and high cognitive loads. Local rates of cerebral glucose metabolism were measured prior to and 9 months after irradiation using [(18)F]-2-deoxy-2-fluoro-d-glucose positron emission tomography. Low cognitive load trials did not reveal a significant reduction in performance until 7 months after irradiation; performance then declined progressively. In high cognitive load trials, the initial impairment was observed ∼1 month after irradiation. This was followed by a transient recovery period over the next 1-2 months, after which performance declined progressively through 11 months after irradiation. Nine months after irradiation, glucose uptake during the DMS task was decreased in the cuneate and prefrontal cortex and was increased in the cerebellum and thalamus compared with the levels prior to irradiation. Results from this pilot study suggest that the radiation-induced changes in cognition and brain metabolism observed in rhesus monkeys may be similar to those observed in brain tumor patients receiving brain irradiation.


Assuntos
Encéfalo/fisiopatologia , Encéfalo/efeitos da radiação , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Lesões por Radiação/fisiopatologia , Animais , Fracionamento da Dose de Radiação , Humanos , Macaca mulatta , Masculino , Projetos Piloto , Doses de Radiação , Lesões por Radiação/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...