Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Med Sci Educ ; 31(1): 15-16, 2021 Feb.
Article in English | MEDLINE | ID: mdl-34457856

ABSTRACT

A case-based laboratory event integrating neuroanatomy, neuroscience, and psychiatry was implemented into a pre-clerkship psychiatry-based course for second-year medical students. Learners rotating through lab stations to work on different cases to make interdisciplinary connections among these fields is an innovative way for them to integrate foundational neurology, neuroanatomy, and psychiatry concepts. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s40670-020-01171-0.

2.
J Neurochem ; 134(5): 833-44, 2015 09.
Article in English | MEDLINE | ID: mdl-26011081

ABSTRACT

The nucleus accumbens is highly heterogeneous, integrating regionally distinct afferent projections and accumbal interneurons, resulting in diverse local microenvironments. Dopamine (DA) neuron terminals similarly express a heterogeneous collection of terminal receptors that modulate DA signaling. Cyclic voltammetry is often used to probe DA terminal dynamics in brain slice preparations; however, this method traditionally requires electrical stimulation to induce DA release. Electrical stimulation excites all of the neuronal processes in the stimulation field, potentially introducing simultaneous, multi-synaptic modulation of DA terminal release. We used optogenetics to selectively stimulate DA terminals and used voltammetry to compare DA responses from electrical and optical stimulation of the same area of tissue around a recording electrode. We found that with multiple pulse stimulation trains, optically stimulated DA release increasingly exceeded that of electrical stimulation. Furthermore, electrical stimulation produced inhibition of DA release across longer duration stimulations. The GABAB antagonist, CGP 55845, increased electrically stimulated DA release significantly more than light stimulated release. The nicotinic acetylcholine receptor antagonist, dihydro-ß-erythroidine hydrobromide, inhibited single pulse electrically stimulated DA release while having no effect on optically stimulated DA release. Our results demonstrate that electrical stimulation introduces local multi-synaptic modulation of DA release that is absent with optogenetically targeted stimulation.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Electric Stimulation , Nucleus Accumbens/cytology , Optogenetics , Presynaptic Terminals/metabolism , Acetylcholine/pharmacology , Animals , Artifacts , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Channelrhodopsins , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Dopaminergic Neurons/drug effects , GABA-B Receptor Antagonists/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Gene Knock-In Techniques , Interneurons/drug effects , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Microelectrodes , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nucleus Accumbens/metabolism , Phosphinic Acids/pharmacology , Presynaptic Terminals/drug effects , Promoter Regions, Genetic , Propanolamines/pharmacology , Synapses/drug effects , Synapses/physiology , Tyrosine 3-Monooxygenase/genetics , Ventral Tegmental Area/metabolism , gamma-Aminobutyric Acid/physiology
3.
Int J Radiat Biol ; 90(9): 799-806, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24991879

ABSTRACT

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.


Subject(s)
Brain/radiation effects , Cognition/radiation effects , Diffusion Tensor Imaging , Dose Fractionation, Radiation , Radiotherapy/methods , Animals , Behavior, Animal/radiation effects , Brain Neoplasms/radiotherapy , Hippocampus/radiation effects , Magnetic Resonance Imaging , Male , Maze Learning , Models, Animal , Random Allocation , Rats , Rats, Inbred F344
4.
Radiat Res ; 181(1): 33-44, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24397438

ABSTRACT

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.


Subject(s)
Cognition/drug effects , Cognition/radiation effects , Fenofibrate/pharmacology , PPAR alpha/agonists , Animals , Behavior, Animal/drug effects , Behavior, Animal/radiation effects , Body Weight/drug effects , Body Weight/radiation effects , Doublecortin Protein , Male , Motor Activity/drug effects , Motor Activity/radiation effects , Rats , Visual Acuity/drug effects , Visual Acuity/radiation effects
5.
Biol Psychiatry ; 74(12): 890-7, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23607969

ABSTRACT

BACKGROUND: The anterior hippocampus is associated with emotional functioning and hippocampal volume is reduced in depression. More women are clinically depressed than men, yet the depressed female brain is little studied. We reported reduced anterior hippocampal volume in behaviorally depressed adult female cynomolgus macaques; the mechanisms contributing to that reduction are unknown. The present study represents the first systematic morphological investigation of the entire hippocampus in depressed female primates. METHODS: Cellular determinants of hippocampal size were examined in subregions of anterior and posterior hippocampus in antidepressant-naïve, adult female monkeys characterized for behavioral depression and matched on variables that influence hippocampal size (n = 8 depressed, 8 nondepressed). Unbiased stereology was used to estimate neuronal and glial numbers, neuronal soma size, and regional and layer volumes. RESULTS: Neuropil and cell layer volumes were reduced in cornu ammonis (CA)1 and dentate gyrus (DG) of the anterior but not the posterior hippocampus of depressed compared with nondepressed monkeys. Glial numbers were 30% lower in anterior CA1 and DG of depressed monkeys, with no differences observed in the posterior hippocampus. Granule neuron number tended toward a reduction in anterior DG; pyramidal neuron number was unchanged in any region. Size of pyramidal neurons and glial densities tended to be reduced throughout the whole hippocampus of depressed monkeys, whereas neuronal densities were unchanged. CONCLUSIONS: The reduced size of the anterior hippocampus in depressed female monkeys appears to arise from alterations in numbers of glia and extent of neuropil, but not numbers of neurons, in CA1 and DG.


Subject(s)
Depression/pathology , Hippocampus/pathology , Neurons/pathology , Neuropil/pathology , Animals , Cell Count , Dexamethasone , Disease Models, Animal , Female , Hierarchy, Social , Hydrocortisone/blood , Macaca fascicularis , Phosphopyruvate Hydratase/metabolism , Progesterone/blood , Statistics, Nonparametric , Stereotaxic Techniques , Steroids/blood
6.
Radiat Res ; 178(1): 46-56, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22687052

ABSTRACT

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.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cerebral Cortex/radiation effects , Cognition Disorders/prevention & control , Cranial Irradiation/adverse effects , Radiation Injuries, Experimental/prevention & control , Ramipril/therapeutic use , Angiotensin I/blood , Animals , Body Weight , Cerebral Cortex/physiology , Cognition Disorders/etiology , Dose Fractionation, Radiation , Doublecortin Protein , Male , Rats , Rats, Inbred F344
7.
PLoS One ; 7(12): e52728, 2012.
Article in English | MEDLINE | ID: mdl-23300752

ABSTRACT

Radiation therapy has proven efficacy for treating brain tumors and metastases. Higher doses and larger treatment fields increase the probability of eliminating neoplasms and preventing reoccurrence, but dose and field are limited by damage to normal tissues. Normal tissue injury is greatest during development and in populations of proliferating cells but also occurs in adults and older individuals and in non-proliferative cell populations. To better understand radiation-induced normal tissue injury and how it may be affected by aging, we exposed young adult, middle-aged, and old rats to 10 Gy of whole brain irradiation and assessed in gray- and white matter the responses of microglia, the primary cellular mediators of radiation-induced neuroinflammation, and oligodendrocyte precursor cells, the largest population of proliferating cells in the adult brain. We found that aging and/or irradiation caused only a few microglia to transition to the classically "activated" phenotype, e.g., enlarged cell body, few processes, and markers of phagocytosis, that is seen following more damaging neural insults. Microglial changes in response to aging and irradiation were relatively modest and three markers of reactivity - morphology, proliferation, and expression of the lysosomal marker CD68- were regulated largely independently within individual cells. Proliferation of oligodendrocyte precursors did not appear to be altered during normal aging but increased following irradiation. The impacts of irradiation and aging on both microglia and oligodendrocyte precursors were heterogeneous between white- and gray matter and among regions of gray matter, indicating that there are regional regulators of the neural response to brain irradiation. By several measures, the CA3 region of the hippocampus appeared to be differentially sensitive to effects of aging and irradiation. The changes assessed here likely contribute to injury following inflammatory challenges like brain irradiation and represent important end-points for analysis in studies of therapeutic strategies to protect patients from neural dysfunction.


Subject(s)
CA3 Region, Hippocampal/pathology , Microglia/radiation effects , Neural Stem Cells/radiation effects , Radiation Injuries, Experimental/pathology , Age Factors , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/pathology , Brain/radiation effects , CA3 Region, Hippocampal/radiation effects , Cell Proliferation , Cell Shape/radiation effects , Male , Microglia/metabolism , Microglia/physiology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Oligodendroglia/metabolism , Oligodendroglia/physiology , Oligodendroglia/radiation effects , Organ Specificity , Phenotype , Rats , Rats, Inbred F344
8.
Radiat Res ; 176(1): 71-83, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21545290

ABSTRACT

Blockers of the renin-angiotensin-aldosterone system (RAAS) ameliorate cognitive deficits and some aspects of brain injury after whole-brain irradiation. We investigated whether treatment with the angiotensin II type 1 receptor antagonist L-158,809 at a dose that protects cognitive function after fractionated whole-brain irradiation reduced radiation-induced neuroinflammation and changes in hippocampal neurogenesis, well-characterized effects that are associated with radiation-induced brain injury. Male F344 rats received L-158,809 before, during and after a single 10-Gy dose of radiation. Expression of cytokines, angiotensin II receptors and angiotensin-converting enzyme 2 was evaluated by real-time PCR 24 h, 1 week and 12 weeks after irradiation. At the latter times, microglial density and proliferating and activated microglia were analyzed in the dentate gyrus of the hippocampus. Cell proliferation and neurogenesis were also quantified in the dentate subgranular zone. L-158,809 treatment modestly increased mRNA expression for Ang II receptors and TNF-α but had no effect on radiation-induced effects on hippocampal microglia or neurogenesis. Thus, although L-158,809 ameliorates cognitive deficits after whole-brain irradiation, the drug did not mitigate the neuroinflammatory microglial response or rescue neurogenesis. Additional studies are required to elucidate other mechanisms of normal tissue injury that may be modulated by RAAS blockers.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Brain/drug effects , Brain/radiation effects , Microglia/cytology , Neurogenesis/drug effects , Neurogenesis/radiation effects , Receptor, Angiotensin, Type 1/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Body Weight/drug effects , Body Weight/radiation effects , Brain/cytology , Brain/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cytokines/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/radiation effects , Drinking/drug effects , Drinking/radiation effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Male , Microglia/drug effects , Microglia/metabolism , Microglia/radiation effects , Peptidyl-Dipeptidase A/genetics , Rats , Rats, Inbred F344 , Receptor, Angiotensin, Type 1/genetics , Whole-Body Irradiation/adverse effects
9.
J Neurol Sci ; 306(1-2): 129-37, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21481894

ABSTRACT

Radiation effectively treats brain tumors and other pathologies but dose and treatment plans are limited by normal tissue injury, a major cause of morbidity in survivors. Clinically significant normal tissue injury can occur even with therapies that target pathological tissue and limit out-of-target irradiation. Elucidating the mechanisms underlying normal tissue injury is facilitated by studying the effects of focal irradiation and comparing irradiated and un-irradiated tissue in experimental animals. Young adult rats were irradiated using the Leksell Gamma Knife® with a 10 Gy maximum dose directed at the left hippocampus and shaped to minimize irradiation contralaterally. At least 95% of targeted hippocampus received ≥3 Gy, while all points in the contralateral hippocampus received <0.3 Gy. Neuronal and microglial markers of damage were assessed in the targeted and contralateral hemispheres of Gamma Knife®-treated rats and compared to non-irradiated controls. Acute cell death and sustained changes in neurogenesis and in microglia occurred in the dentate gyrus of the targeted, but not the contralateral, hippocampus, providing experimental evidence that focal irradiation at doses received by peri-target regions during targeted radiation therapy produces robust normal tissue responses. Additional studies using this approach will facilitate assessment of in vivo dose responses and the cellular and molecular mechanisms of radiation-induced brain injury.


Subject(s)
Gamma Rays , Gene Expression Regulation/radiation effects , Hippocampus/pathology , Hippocampus/radiation effects , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Differentiation/radiation effects , Dose-Response Relationship, Radiation , Ectodysplasins/metabolism , Functional Laterality/radiation effects , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/metabolism , Microglia/radiation effects , Neurogenesis/radiation effects , Rats , Rats, Inbred F344 , Stereotaxic Techniques , Time Factors
10.
J Gerontol A Biol Sci Med Sci ; 66(5): 521-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21393424

ABSTRACT

Fish oil (FO) mediates a number of cardioprotective benefits in patients with cardiovascular disease. In the absence of cardiovascular disease, however, the effects of FO on cardiac structure and function are not clear. In addition, it is not known if an effective dosing strategy for attenuating age-related cardiac dysfunction is also effective at limiting cognitive dysfunction. Therefore, we determined if 4 months of FO supplementation in aged rats would lessen age-related cardiac dysfunction while concomitantly preventing the cognitive decline that is normally observed in this population. The results indicate that FO initiated late in life modifies diastolic function in a small but positive way by attenuating the age-related increases in filling pressure, posterior wall thickness, and interstitial collagen without mitigating age-related deficits in memory or increases in brain inflammation. These data raise the possibility that FO supplementation for purposes of cardiac and brain protection may need to occur earlier in the life span.


Subject(s)
Aging/physiology , Brain/pathology , Diastole/drug effects , Dietary Fats, Unsaturated/pharmacology , Fish Oils/pharmacology , Memory/drug effects , Aging/drug effects , Animals , Body Weight/drug effects , Brain/drug effects , Cell Count , Cognition/drug effects , Cognition/physiology , Diastole/physiology , Encephalitis , Fatty Acids, Omega-3/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Immunohistochemistry , Male , Motor Activity/drug effects , Motor Activity/physiology , Organ Size/drug effects , Rats , Vision, Ocular/drug effects , Vision, Ocular/physiology
11.
Neurobiol Aging ; 32(12): 2322.e1-4, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20561717

ABSTRACT

Degeneration of the cholinergic neurons in the basal forebrain and elevation of inflammatory markers are well-established hallmarks of Alzheimer's disease; however, the interplay of these processes in normal aging is not extensively studied. Consequently, we conducted a neuroanatomical investigation to quantify cholinergic neurons and activated microglia in the medial septum/vertical diagonal band (MS/VDB) of young (6 months) and aged (28 months) Fisher 344 × Brown Norway F(1) rats. Aged rats in this study were impaired relative to the young animals in spatial learning ability as assessed in the Morris water maze. Stereological analysis revealed no difference between aged and young rats in the total numbers of cholinergic neurons, demonstrating that loss of cholinergic neurons is not a necessary condition to observe impaired spatial learning in aged rats. In this same region, the total number of activated microglia was substantially greater in aged rats relative to young rats. Jointly, these data demonstrate that aging is characterized by an increase in the basal inflammatory state within the MS/VDB, but this inflammation is not associated with cholinergic neuron death.


Subject(s)
Aging , Brain Chemistry , Cholinergic Neurons/chemistry , Nerve Degeneration , Aging/pathology , Animals , Cholinergic Neurons/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Nerve Degeneration/pathology , Rats , Rats, Inbred BN , Rats, Inbred F344
12.
Radiat Res ; 173(1): 49-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20041759

ABSTRACT

Cognitive dysfunction develops in approximately 50% of patients who receive fractionated whole-brain irradiation and survive 6 months or more. The mechanisms underlying these deficits are unknown. A recent study demonstrated that treatment with the angiotensin II type 1 receptor antagonist (AT(1)RA) L-158,809 before, during and after fractionated whole-brain irradiation prevents or ameliorates radiation-induced cognitive deficits in adult rats. Given that (1) AT(1)RAs may function as anti-inflammatory drugs, (2) inflammation is thought to contribute to radiation injury, and (3) radiation-induced inflammation alters progenitor cell populations, we tested whether the cognitive benefits of L-158,809 treatment were associated with amelioration of the sustained neuroinflammation and changes in neurogenesis that are induced by fractionated whole-brain irradiation. In rats examined 28 and 54 weeks after irradiation, L-158,809 treatment did not alter the effects of radiation on the number and activation of microglia in the perirhinal cortex and hippocampus, nor did it prevent the radiation-induced decrease in proliferating cells and immature neurons in the hippocampus. These findings suggest that L-158,809 does not prevent or ameliorate radiation-induced cognitive deficits by modulation of chronic inflammatory mechanisms, but rather may reduce radiation-induced changes that occur earlier in the postirradiation period and that lead to cognitive dysfunction.


Subject(s)
Brain/radiation effects , Imidazoles/pharmacology , Microglia/drug effects , Microglia/radiation effects , Neurogenesis/drug effects , Neurogenesis/radiation effects , Receptor, Angiotensin, Type 1/metabolism , Tetrazoles/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/radiation effects , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Dentate Gyrus/radiation effects , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Microglia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurons/radiation effects , Radiation Dosage , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Rats , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/radiation effects , Time Factors
13.
PPAR Res ; 2010: 234975, 2010.
Article in English | MEDLINE | ID: mdl-19789638

ABSTRACT

Whole-brain irradiation (WBI) represents the primary mode of treatment for brain metastases; about 200 000 patients receive WBI each year in the USA. Up to 50% of adult and 100% of pediatric brain cancer patients who survive >6 months post-WBI will suffer from a progressive, cognitive impairment. At present, there are no proven long-term treatments or preventive strategies for this significant radiation-induced late effect. Recent studies suggest that the pathogenesis of radiation-induced brain injury involves WBI-mediated increases in oxidative stress and/or inflammatory responses in the brain. Therefore, anti-inflammatory strategies can be employed to modulate radiation-induced brain injury. Peroxisomal proliferator-activated receptors (PPARs) are ligand-activated transcription factors that belong to the steroid/thyroid hormone nuclear receptor superfamily. Although traditionally known to play a role in metabolism, increasing evidence suggests a role for PPARs in regulating the response to inflammation and oxidative injury. PPAR agonists have been shown to cross the blood-brain barrier and confer neuroprotection in animal models of CNS disorders such as stroke, multiple sclerosis and Parkinson's disease. However, the role of PPARs in radiation-induced brain injury is unclear. In this manuscript, we review the current knowledge and the emerging insights about the role of PPARs in modulating radiation-induced brain injury.

14.
Int J Radiat Oncol Biol Phys ; 75(3): 870-7, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19801103

ABSTRACT

PURPOSE: Whole-brain irradiation (WBI) leads to cognitive impairment months to years after radiation. Numerous studies suggest that decreased hippocampal neurogenesis and microglial activation are involved in the pathogenesis of WBI-induced brain injury. The goal of this study was to investigate whether administration of the peroxisomal proliferator-activated receptor (PPAR) alpha agonist fenofibrate would prevent the detrimental effect of WBI on hippocampal neurogenesis. METHODS AND MATERIALS: For this study, 129S1/SvImJ wild-type and PPARalpha knockout mice that were fed either regular or 0.2% wt/wt fenofibrate-containing chow received either sham irradiation or WBI (10-Gy single dose of (137)Cs gamma-rays). Mice were injected intraperitoneally with bromodeoxyuridine to label the surviving cells at 1 month after WBI, and the newborn neurons were counted at 2 months after WBI by use of bromodeoxyuridine/neuronal nuclei double immunofluorescence. Proliferation in the subgranular zone and microglial activation were measured at 1 week and 2 months after WBI by use of Ki-67 and CD68 immunohistochemistry, respectively. RESULTS: Whole-brain irradiation led to a significant decrease in the number of newborn hippocampal neurons 2 months after it was performed. Fenofibrate prevented this decrease by promoting the survival of newborn cells in the dentate gyrus. In addition, fenofibrate treatment was associated with decreased microglial activation in the dentate gyrus after WBI. The neuroprotective effects of fenofibrate were abolished in the knockout mice, indicating a PPARalpha-dependent mechanism or mechanisms. CONCLUSIONS: These data highlight a novel role for PPARalpha ligands in improving neurogenesis after WBI and offer the promise of improving the quality of life for brain cancer patients receiving radiotherapy.


Subject(s)
Cranial Irradiation/adverse effects , Fenofibrate/pharmacology , Hippocampus/cytology , Microglia/drug effects , Neurogenesis/drug effects , PPAR alpha/agonists , Animals , Brain/radiation effects , Bromodeoxyuridine/administration & dosage , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cognition Disorders/etiology , Cognition Disorders/prevention & control , Mice , Mice, Knockout , Microglia/radiation effects , Neurogenesis/radiation effects , Radiation-Protective Agents/pharmacology
15.
Growth Factors ; 27(3): 181-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19343576

ABSTRACT

Insulin-like growth factor-I (IGF-I), a functionally important neurotrophic factor, impacts tissues throughout the body including the central nervous system. In addition to the significant proportion of IGF-I that is synthesized in the liver and released into the plasma, IGF-I is expressed locally in tissues. The present study investigated the relationship between plasma and local brain levels of IGF-I in two well-characterized models of decreased IGF-I. The first is an adult-onset growth hormone deficiency (AOGHD) model, and the second is a caloric restriction (CR) model. In the first cohort of animals from both models, the hippocampus was removed from the brain immediately following decapitation, and in the second cohort, the animals were perfused transcardially with phosphate buffered saline to remove cerebral blood prior to harvesting the hippocampus. Our results demonstrated that although the plasma IGF-I levels were decreased in the CR and AOGHD rats compared to controls, the hippocampal IGF-I levels did not differ among the groups. These data suggest that local brain IGF-I levels are regulated in a different manner than plasma IGF-I levels.


Subject(s)
Dwarfism/metabolism , Hippocampus/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Caloric Restriction , Hippocampus/blood supply , Male , Rats
16.
Glia ; 57(10): 1062-71, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19115393

ABSTRACT

Growth hormone (GH) and insulin-like growth factor-I (IGF-I) provide trophic support during development and also appear to influence cell structure, function and replacement in the adult brain. Recent studies demonstrated effects of the GH/IGF-I axis on adult neurogenesis, but it is unclear whether the GH/IGF-I axis influences glial turnover in the normal adult brain. In the current study, we used a selective model of adult-onset GH and IGF-I deficiency to evaluate the role of GH and IGF-I in regulating glial proliferation and survival in the adult corpus callosum. GH/IGF-I-deficient dwarf rats of the Lewis strain were made GH/IGF-I replete via twice daily injections of GH starting at postnatal day 28 (P28), approximately the age at which GH pulse amplitude increases in developing rodents. GH/IGF-I deficiency was initiated in adulthood by removing animals from GH treatment. Quantitative analyses revealed that adult-onset GH/IGF-I deficiency decreased cell proliferation in the white matter and decreased the survival of newborn oligodendrocytes. These findings are consistent with the hypothesis that aging-related changes in the GH/IGF-I axis produce deficits in ongoing turnover of oligodendrocytes, which may contribute to aging-related cognitive changes and deficits in remyelination after injury.


Subject(s)
Aging/metabolism , Cell Proliferation , Corpus Callosum/metabolism , Growth Hormone/deficiency , Insulin-Like Growth Factor I/deficiency , Oligodendroglia/metabolism , Aging/pathology , Animals , Cell Survival/physiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Disease Models, Animal , Female , Growth Hormone/pharmacology , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Memory Disorders/physiopathology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Nerve Regeneration/physiology , Oligodendroglia/pathology , Rats , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/physiopathology
17.
Exp Neurol ; 211(1): 141-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18342310

ABSTRACT

Caloric restriction (CR) is a daily reduction of total caloric intake without a decrease in micronutrients or disproportionate reduction of any one dietary component. CR can increase lifespan reliably in a wide range of species and appears to counteract some aspects of the aging process throughout the body. The effects on the brain are less clear, but moderate CR seems to attenuate age-related cognitive decline. Thus, we determined the effects of age and CR on key synaptic proteins in the CA3 region of the hippocampus and whether these changes were correlated with differences in behavior on a hippocampal-dependent learning and memory task. We observed an overall, age-related decline in the NR1, N2A and N2B subunits of the N-methyl-d-aspartate (NMDA)-type and the GluR1 and GluR2 subunits of the alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionic acid (AMPA)-type ionotropic glutamate receptors. Interestingly, we found that CR initially lowers the glutamate receptor subunit levels as compared to young AL animals, and then stabilizes the levels across lifespan. Synaptophysin, a presynaptic vesicle protein, showed a similar pattern. We also found that both CR and ad libitum (AL) fed animals exhibited age-related cognitive decline on the Morris water maze task. However, AL animals declined between young and middle age, and between middle age and old, whereas CR rats only declined between young and middle age. Thus, the decrease in key synaptic proteins in CA3 and cognitive decline occurring across lifespan are stabilized by CR. This age-related decrease and CR-induced stabilization are likely to affect CA3 synaptic plasticity and, as a result, hippocampal function.


Subject(s)
Aging/physiology , Caloric Restriction , Hippocampus/physiology , Learning/physiology , Receptors, Glutamate/metabolism , Space Perception/physiology , Age Factors , Analysis of Variance , Animals , Behavior, Animal , Gene Expression Regulation , Male , Maze Learning , Rats , Receptors, Glutamate/classification , Swimming
18.
J Gerontol A Biol Sci Med Sci ; 63(1): 12-20, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18245756

ABSTRACT

The effect of aging on microvascular density and plasticity in the rodent hippocampus, a brain region critically important for learning and memory, was investigated in F344xBN rats. Capillary density and angiogenesis were measured in three regions of the hippocampus in young and old rats and in old rats administered growth hormone, a treatment that improves cognitive function in older animals. Animals were housed under control conditions or in hypoxic conditions (11% ambient oxygen levels) to stimulate vascular growth. Our results indicate that aging is not associated with a reduction in hippocampal capillary density. However, aged animals demonstrate a significant impairment in hypoxia-induced capillary angiogenesis compared to young animals. Growth hormone treatment to aged animals for 6 weeks did not alter hippocampal capillary density and did not ameliorate the age-related deficit in angiogenesis. We conclude that aging significantly reduces hippocampal microvascular plasticity, which is not improved with growth hormone therapy.


Subject(s)
Aging/physiology , Hippocampus/physiopathology , Hypoxia/complications , Neovascularization, Physiologic/physiology , Animals , Growth Hormone/administration & dosage , Hippocampus/physiology , Models, Animal , Neovascularization, Physiologic/drug effects , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
19.
Int J Radiat Oncol Biol Phys ; 70(3): 826-34, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18164853

ABSTRACT

PURPOSE: To assess the impact of aging on the radiation response in the adult rat brain. METHODS AND MATERIALS: Male rats 8, 18, or 28 months of age received a single 10-Gy dose of whole-brain irradiation (WBI). The hippocampal dentate gyrus was analyzed 1 and 10 weeks later for sensitive neurobiologic markers associated with radiation-induced damage: changes in density of proliferating cells, immature neurons, total microglia, and activated microglia. RESULTS: A significant decrease in basal levels of proliferating cells and immature neurons and increased microglial activation occurred with normal aging. The WBI induced a transient increase in proliferation that was greater in older animals. This proliferation response did not increase the number of immature neurons, which decreased after WBI in young rats, but not in old rats. Total microglial numbers decreased after WBI at all ages, but microglial activation increased markedly, particularly in older animals. CONCLUSIONS: Age is an important factor to consider when investigating the radiation response of the brain. In contrast to young adults, older rats show no sustained decrease in number of immature neurons after WBI, but have a greater inflammatory response. The latter may have an enhanced role in the development of radiation-induced cognitive dysfunction in older individuals.


Subject(s)
Aging/physiology , Brain/radiation effects , Microglia/radiation effects , Neurons/radiation effects , Age Factors , Animals , Brain/pathology , Brain/physiology , Cell Count , Cell Proliferation/radiation effects , Cranial Irradiation/methods , Dentate Gyrus/chemistry , Dentate Gyrus/pathology , Dentate Gyrus/radiation effects , Male , Microglia/cytology , Neurons/cytology , Rats , Rats, Inbred F344
20.
Neurobiol Aging ; 29(9): 1308-18, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17433502

ABSTRACT

Caloric restriction (CR) attenuates aging-related degenerative processes throughout the body. It is less clear, however, whether CR has a similar effect in the brain, particularly in the hippocampus, an area important for learning and memory processes that often are compromised in aging. In order to evaluate the effect of CR on synapses across lifespan, we quantified synapses stereologically in the middle molecular layer of the dentate gyrus (DG) of young, middle aged and old Fischer 344 x Brown Norway rats fed ad libitum (AL) or a CR diet from 4 months of age. The results indicate that synapses are maintained across lifespan in both AL and CR rats. In light of this stability, we addressed whether aging and CR influence neurotransmitter receptor levels by measuring subunits of NMDA (NR1, NR2A and NR2B) and AMPA (GluR1, GluR2) receptors in the DG of a second cohort of AL and CR rats across lifespan. The results reveal that the NR1 and GluR1 subunits decline with age in AL, but not CR rats. The absence of an aging-related decline in these subunits in CR rats, however, does not arise from increased levels in old CR rats. Instead, it is due to subunit decreases in young CR rats to levels that are sustained in CR rats throughout lifespan, but that are reached in AL rats only in old age.


Subject(s)
Aging/metabolism , Aging/pathology , Caloric Restriction/methods , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synapses/pathology , Animals , Humans , Male , Models, Animal , Protein Subunits/metabolism , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL