Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 254
Filter
1.
J Prev Alzheimers Dis ; 11(3): 537-548, 2024.
Article in English | MEDLINE | ID: mdl-38706270

ABSTRACT

BACKGROUND: Monoclonal antibodies that target amyloid-beta and remove amyloid plaques can slow cognitive and functional decline in early Alzheimer's disease. Gantenerumab is a subcutaneously administered fully-human anti-amyloid-beta monoclonal antibody with highest affinity for aggregated amyloid-beta. Since the phase 3 GRADUATE trials did not meet the primary endpoint (change from baseline to Week 116 in Clinical Dementia Rating scale - Sum of Boxes), development of gantenerumab in sporadic Alzheimer's disease was stopped and all ongoing trials were terminated early due to sponsor decision. Subcutaneous administration at the clinic or at home by care partner would be an important option for other therapies in this class in order to increase flexibility and reduce overall burden. The insights obtained from the experience with gantenerumab home administration by care partner in the phase 2 GRADUATION trial will serve to guide the ongoing efforts with other anti-amyloid-beta antibodies. OBJECTIVES: To evaluate the pharmacodynamic effects on brain amyloid load of once weekly subcutaneous administration of gantenerumab and the safety and feasibility of home administration by care partners. DESIGN: Phase 2, open-label, single arm study. SETTING: Multicenter trial conducted in 33 sites in 8 countries from November 2020 to March 2023. PARTICIPANTS: Participants aged 50 to 90 with early symptomatic Alzheimer's disease (mild cognitive impairment/mild dementia due to Alzheimer's disease), and evidence of amyloid positron emission tomography positivity. INTERVENTION: Participants could receive up to 255 mg gantenerumab once-weekly, administered subcutaneously at site or at home by healthcare professionals or non-healthcare-professional care partners. MEASUREMENTS: The primary endpoint was the change from baseline to Week 52 and to Week 104 in brain amyloid load as measured by PET centiloid levels. The secondary endpoints were responses to the home administration questionnaire, plasma concentrations and safety. RESULTS: The overall number of participants enrolled was 192, with a mean (standard deviation) amyloid PET load at baseline of 101.80 (29.80) centiloids. At the time of early study termination by sponsor, 149 participants had valid Week 52 amyloid PET data (primary endpoint), and 12 participants had an early termination PET within the pre-defined time range of Week 104. The mean change in amyloid PET from baseline to Week 52 and Week 104 was -26.19 centiloids (range: -75.6-15.8; n=149) and -35.48 centiloids (range: -63.2--7.0; n=12), respectively. Responses to the home administration questionnaire at Week 52 (n=148) indicated that the majority of care partners (88-97%) considered administration of study drug at home easy (30.4%) or very easy (57.4%), and convenient (25.7%) or very convenient (70.9%). Care partners felt confident (31.1%) or very confident (62.2%) and satisfied (29.7%) or very satisfied (64.9%) with giving the injection at home. Responses by care partners at Week 36 (n=72), Week 76 (n=126) and Week 104 (n=29) and participant (patient) assessment of convenience and satisfaction at these time points were similar. There were no new safety findings associated with gantenerumab administered subcutaneously once weekly at 255 mg or safety issues associated with at-home injections by non-healthcare professional care partners. CONCLUSIONS: Once-weekly subcutaneous home administration of the anti-amyloid-beta antibody gantenerumab by non-healthcare-professional care partners to participants with early Alzheimer's disease was feasible, safe, well tolerated, and considered as a convenient option by both the care partners and participants with Alzheimer's disease. Although gantenerumab's development has been stopped due to lack of efficacy, this approach has the potential to reduce the frequency of hospital/outpatient clinic visits required for treatment with other anti-amyloid-ß antibodies and can increase flexibility of drug administration for people living with Alzheimer's disease and their families.


Subject(s)
Alzheimer Disease , Antibodies, Monoclonal, Humanized , Feasibility Studies , Humans , Alzheimer Disease/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Aged , Female , Male , Caregivers , Positron-Emission Tomography , Amyloid beta-Peptides/metabolism , Injections, Subcutaneous , Brain/drug effects , Brain/metabolism , Brain/diagnostic imaging , Middle Aged , Aged, 80 and over
2.
Hippocampus ; 28(10): 745-764, 2018 10.
Article in English | MEDLINE | ID: mdl-29989271

ABSTRACT

The dynamic process of memory consolidation involves a reorganization of brain regions that support a memory trace over time, but exactly how the network reorganizes as the memory changes remains unclear. We present novel converging evidence from studies of animals (rats) and humans for the time-dependent reorganization and transformation of different types of memory as measured both by behavior and brain activation. We find that context-specific memories in rats, and naturalistic episodic memories in humans, lose precision over time and activity in the hippocampus decreases. If, however, the retrieved memories retain contextual or perceptual detail, the hippocampus is engaged similarly at recent and remote timepoints. As the interval between the timepoint increases, the medial prefrontal cortex is engaged increasingly during memory retrieval, regardless of the context or the amount of retrieved detail. Moreover, these hippocampal-frontal shifts are accompanied by corresponding changes in a network of cortical structures mediating perceptually-detailed as well as less precise, schematic memories. These findings provide cross-species evidence for the crucial interplay between hippocampus and neocortex that reflects changes in memory representation over time and underlies systems consolidation.


Subject(s)
Brain/physiology , Hippocampus/cytology , Memory Consolidation/physiology , Memory, Episodic , Neurons/physiology , Adult , Analysis of Variance , Animals , Avoidance Learning/physiology , Brain/diagnostic imaging , Fear/psychology , Female , Functional Laterality , Hippocampus/diagnostic imaging , Hippocampus/physiology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Oxygen/blood , Proto-Oncogene Proteins c-fos/metabolism , Random Allocation , Rats , Rats, Long-Evans , Time Factors , Young Adult
3.
J Alzheimers Dis ; 64(3): 835-857, 2018.
Article in English | MEDLINE | ID: mdl-29914019

ABSTRACT

The increasing global burden of Alzheimer's disease (AD) and failure of conventional treatments to stop neurodegeneration necessitates an alternative approach. Evidence of inflammation, mitochondrial dysfunction, and oxidative stress prior to the accumulation of amyloid-ß in the prodromal stage of AD (mild cognitive impairment; MCI) suggests that early interventions which counteract these features, such as dietary supplements, may ameliorate the onset of MCI-like behavioral symptoms. We administered a polyphenol-containing multiple ingredient dietary supplement (MDS), or vehicle, to both sexes of triple transgenic (3xTg-AD) mice and wildtype mice for 2 months from 2-4 months of age. We hypothesized that the MDS would preserve spatial learning, which is known to be impaired in untreated 3xTg-AD mice by 4 months of age. Behavioral phenotyping of animals was done at 1-2 and 3-4 months of age using a comprehensive battery of tests. As previously reported in males, both sexes of 3xTg-AD mice exhibited increased anxiety-like behavior at 1-2 months of age, prior to deficits in learning and memory, which did not appear until 3-4 months of age. The MDS did not reduce this anxiety or prevent impairments in novel object recognition (both sexes) or on the water maze probe trial (females only). Strikingly, the MDS specifically prevented 3xTg-AD mice (both sexes) from developing impairments (exhibited by untreated 3xTg-AD controls) in working memory and spatial learning. The MDS also increased sucrose preference, an indicator of hedonic tone. These data show that the MDS can prevent some, but not all, psychopathology in an AD model.


Subject(s)
Alzheimer Disease/complications , Dietary Supplements , Memory Disorders/diet therapy , Memory Disorders/etiology , Mood Disorders/diet therapy , Mood Disorders/etiology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Disease Models, Animal , Exploratory Behavior/physiology , Female , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Strength/drug effects , Mutation/genetics , Presenilin-1/genetics , Psychomotor Performance/physiology , Smell/physiology , tau Proteins/genetics
4.
J Neurogenet ; 32(3): 142-146, 2018 09.
Article in English | MEDLINE | ID: mdl-29790805

ABSTRACT

Twenty years spent in one laboratory is sufficient to build a legacy of publications and a body of work to make an impact. However, the impact of our work was highest at the personal level, and time spent in Harold Atwood's laboratory was not a culmination of my career but rather a crucial path toward learning and maturing as a researcher. During that time, I experienced discoveries and lessons that shaped the next steps of my career. This article is written in gratitude for wonderful experiences and describes a few highlights that were especially memorable and influential.


Subject(s)
Neurology/history , History, 20th Century , History, 21st Century
5.
PLoS One ; 13(1): e0191866, 2018.
Article in English | MEDLINE | ID: mdl-29370277

ABSTRACT

Chemotherapy-induced cognitive impairment (CICI) occurs in a substantial proportion of treated cancer patients, with no drug currently available for its therapy. This study investigated whether PAN-811, a ribonucleotide reductase inhibitor, can reduce cognitive impairment and related suppression of neurogenesis following chemotherapy in an animal model. Young adult rats in Chemo and Chemo+PAN-811 groups received 3 intraperitoneal (i.p.) injections of methotrexate (MTX) and 5-fluorouracil (5-FU), and those in Saline and Saline+PAN-811 groups received equal volumes of physiological saline at 10-day intervals. PAN-811 in saline was delivered through i.p. injection, 10 min following each saline (Saline+PAN-811 group) or MTX/5-FU (Chemo+PAN-811 group) treatment, while equal volumes of saline were delivered to Saline and Chemo groups. Over Days 31-66, rats were administered tests of spatial memory, nonmatching-to-sample rule learning, and discrimination learning, which are sensitive to dysfunction in hippocampus, frontal lobe and striatum, respectively. On Day 97, neurogenesis was immnunohistochemically evaluated by counting doublecortin-positive (DCX+) cells in the dentate gyrus (DG). The results demonstrated that the Chemo group was impaired on the three cognitive tasks, but co-administration of PAN-811 significantly reduced all MTX/5-FU-induced cognitive impairments. In addition, MTX/5-FU reduced DCX+ cells to 67% of that in Saline control rats, an effect that was completely blocked by PAN-811 co-administration. Overall, we present the first evidence that PAN-811 protects cognitive functions and preserves neurogenesis from deleterious effects of MTX/5-FU. The current findings provide a basis for rapid clinical translation to determine the effect of PAN-811 on CICI in human.


Subject(s)
Cognitive Dysfunction/prevention & control , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Pyridines/pharmacology , Thiosemicarbazones/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cognitive Dysfunction/chemically induced , Dentate Gyrus/drug effects , Discrimination Learning/drug effects , Disease Models, Animal , Doublecortin Protein , Enzyme Inhibitors/pharmacology , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Learning/drug effects , Methotrexate/administration & dosage , Methotrexate/adverse effects , Rats , Rats, Long-Evans , Ribonucleotide Reductases/antagonists & inhibitors , Spatial Memory/drug effects
6.
Neuroscience ; 369: 51-65, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29113931

ABSTRACT

Animal studies have reinforced clinical reports of cognitive impairment in cancer survivors following chemotherapy but, until now, all pre-clinical research in this area has been conducted on normal rodents. The present study investigated the effects of chemotherapy on cognition and underlying biological mechanisms in the FVB/N-Tg (MMTV-neu) 202 Mul/J mouse, a well-characterized transgenic model of breast cancer that has similarities to the tumorigenesis which occurs in humans. Tumor-bearing and control mice received three weekly injections of a combination of methotrexate + 5-fluorouracil, or an equal volume of saline. Different aspects of learning and memory were measured before and after treatment. The effects of tumor and chemotherapy on neurogenesis, neuro-inflammatory cytokine activity, and brain volume, as they relate to corresponding cognitive changes, were also measured. The toxic effects of chemotherapy extended to the cancerous model in which substantial cognitive impairment was also associated with the disease. Cognitive deficits were greatest in tumorigenic mice that received the anti-cancer drugs. Both tumor growth and chemotherapy caused significant changes in brain volume, including the hippocampus and frontal lobes, two structures that are directly implicated in cognitive tasks that were shown to be vulnerable. The level of hippocampal neurogenesis in adulthood was suppressed in chemotherapy-treated mice and associated with loss of hippocampus-controlled cognitive function. Dysregulation of cytokine activity was found in tumorigenic mice and associated with impaired cognitive performance. The results show that chemotherapy and tumor development independently contribute to cognitive deficits through different biological mechanisms.


Subject(s)
Breast Neoplasms/psychology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/psychology , Fluorouracil/adverse effects , Methotrexate/adverse effects , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain/drug effects , Brain/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cytokines/metabolism , Disease Models, Animal , Female , Learning/drug effects , Memory/drug effects , Mice , Mice, Transgenic , Neurogenesis/drug effects , Organ Size/drug effects , Receptors, Virus/genetics
7.
eNeuro ; 4(4)2017.
Article in English | MEDLINE | ID: mdl-28824956

ABSTRACT

Cognitive reserve, the brain's capacity to draw on enriching experiences during youth, is believed to protect against memory loss associated with a decline in hippocampal function, as seen in normal aging and neurodegenerative disease. Adult neurogenesis has been suggested as a specific mechanism involved in cognitive (or neurogenic) reserve. The first objective of this study was to compare learning-related neuronal activity in adult-born versus developmentally born hippocampal neurons in juvenile male rats that had engaged in extensive running activity during early development or reared in a standard laboratory environment. The second objective was to investigate the long-term effect of exercise in rats on learning and memory of a contextual fear (CF) response later in adulthood. These aims address the important question as to whether exercise in early life is sufficient to build a reserve that protects against the process of cognitive aging. The results reveal a long-term effect of early running on adult-born dentate granule neurons and a special role for adult-born neurons in contextual memory, in a manner that is consistent with the neurogenic reserve hypothesis.


Subject(s)
Aging/physiology , Dentate Gyrus/cytology , Fear/physiology , Learning/physiology , Neurons/physiology , Physical Conditioning, Animal/physiology , Analysis of Variance , Animals , Cell Count , Dentate Gyrus/physiology , Gene Expression Regulation/physiology , Male , Memory/physiology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Long-Evans , Running
8.
Behav Neurosci ; 130(4): 428-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27337062

ABSTRACT

Clinical studies indicate that up to 70% of cancer patients who receive chemotherapy experience cognitive impairment. The present study investigated environmental enrichment as a protective factor against the adverse effects of anticancer drugs on cognitive and biological processes in an animal model. Adult rats were housed in group cages with environmental stimulation or in standard cages for 3 months, before receiving 3 weekly injections of methotrexate + 5-fluorouracil, or equal volumes of saline. Rats were then administered tests of learning and memory that are sensitive to hippocampal or frontal lobe dysfunction. The relationship between cognitive performance and hippocampal neurogenesis was examined through sensitive time-dependent measures of neuronal maturation. Chemotherapy-treated rats in the standard environment were impaired on tests of spatial memory, nonmatching-to-sample (NMTS) rule learning, and delayed-NMTS. Chemotherapy-treated rats in the enriched environment performed at or near normal levels. The performance of the chemotherapy groups on the hippocampus-sensitive, spatial memory and delayed-NMTS tests correlated with neurogenesis levels. The results show that environmental enrichment can reduce the risk of chemotherapy-induced cognitive impairment, in part by promoting neuronal differentiation and growth during cell maturation. As well, they point to the importance of lifestyle factors in treating or preventing adverse effects of anticancer drugs on cognitive function. (PsycINFO Database Record


Subject(s)
Cognition Disorders/chemically induced , Disease Models, Animal , Drug Therapy/methods , Environment , Animals , Antimetabolites, Antineoplastic/adverse effects , Cognition Disorders/prevention & control , Female , Fluorouracil/adverse effects , Hippocampus , Methotrexate/adverse effects , Neurogenesis/drug effects , Neurogenesis/physiology , Rats , Rats, Long-Evans , Spatial Memory/drug effects , Spatial Memory/physiology
9.
Neuroscience ; 308: 180-93, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26358368

ABSTRACT

Severe chronic stress can have a profoundly negative impact on the brain, affecting plasticity, neurogenesis, memory and mood. On the other hand, there are factors that upregulate neurogenesis, which include dietary antioxidants and physical activity. These factors are associated with biochemical processes that are also altered in age-related cognitive decline and dementia, such as neurotrophin expression, oxidative stress and inflammation. We exposed mice to an unpredictable series of stressors or left them undisturbed (controls). Subsets of stressed and control mice were concurrently given (1) no additional treatment, (2) a complex dietary supplement (CDS) designed to ameliorate inflammation, oxidative stress, mitochondrial dysfunction, insulin resistance and membrane integrity, (3) a running wheel in each of their home cages that permitted them to exercise, or (4) both the CDS and the running wheel for exercise. Four weeks of unpredictable stress reduced the animals' preference for saccharin, increased their adrenal weights and abolished the exercise-induced upregulation of neurogenesis that was observed in non-stressed animals. Unexpectedly, stress did not reduce hippocampal size, brain-derived neurotrophic factor (BDNF), or neurogenesis. The combination of dietary supplementation and exercise had multiple beneficial effects, as reflected in the number of doublecortin (DCX)-positive immature neurons in the dentate gyrus (DG), the sectional area of the DG and hippocampal CA1, as well as increased hippocampal BDNF messenger ribonucleic acid (mRNA) and serum vascular endothelial growth factor (VEGF) levels. In contrast, these benefits were not observed in chronically stressed animals exposed to either dietary supplementation or exercise alone. These findings could have important clinical implications for those suffering from chronic stress-related disorders such as major depression.


Subject(s)
Dietary Supplements , Hippocampus/physiopathology , Running/physiology , Stress, Psychological/physiopathology , Stress, Psychological/therapy , Animals , Brain-Derived Neurotrophic Factor/metabolism , Chronic Disease , Depressive Disorder/pathology , Depressive Disorder/physiopathology , Depressive Disorder/therapy , Diet , Disease Models, Animal , Doublecortin Protein , Hippocampus/pathology , Insulin-Like Growth Factor I/metabolism , Male , Mice, Inbred C57BL , Neurogenesis/physiology , Organ Size , Physical Conditioning, Animal/physiology , Stress, Psychological/pathology , Treatment Outcome , Uncertainty , Vascular Endothelial Growth Factor A/blood
10.
J Vis Exp ; (97)2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25867138

ABSTRACT

Here we describe a technique for studying hippocampal postnatal neurogenesis in the rodent brain using the organotypic slice culture technique. This method maintains the characteristic topographical morphology of the hippocampus while allowing direct application of pharmacological agents to the developing hippocampal dentate gyrus. Additionally, slice cultures can be maintained for up to 4 weeks and thus, allow one to study the maturation process of newborn granule neurons. Slice cultures allow for efficient pharmacological manipulation of hippocampal slices while excluding complex variables such as uncertainties related to the deep anatomic location of the hippocampus as well as the blood brain barrier. For these reasons, we sought to optimize organotypic slice cultures specifically for postnatal neurogenesis research.


Subject(s)
Dentate Gyrus/cytology , Hippocampus/cytology , Neurogenesis/physiology , Neurons/cytology , Organ Culture Techniques/methods , Animals , Hippocampus/growth & development , Rats , Rats, Sprague-Dawley
11.
Behav Brain Res ; 281: 239-44, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25529185

ABSTRACT

Drugs used to treat cancer have neurotoxic effects that often produce memory loss and related cognitive deficits. In a test of the hypothesis that chemotherapy-induced cognitive impairment is related to a loss of inhibitory control, rats injected with a combination of methotrexate+5-fluouracil or equal volumes of saline, were administered a retroactive interference task in which memory for a learned discrimination problem was tested under conditions of high- and low-interference. The drugs had no effect on original learning or on re-learning the discrimination response when there was little interference, but the chemotherapy group was severely impaired in the hippocampus-sensitive, high-interference memory test. The impaired performance correlated significantly with reduced neurogenesis in the hippocampus. The failure to suppress interfering influences is consistent with a breakdown in pattern separation, a process that distinguishes and separates overlapping neural representations of experiences that have a high degree of similarity.


Subject(s)
Antineoplastic Agents/adverse effects , Hippocampus/pathology , Memory Disorders/chemically induced , Memory/drug effects , Neurogenesis/drug effects , Psychomotor Performance/drug effects , Animals , Antimetabolites, Antineoplastic/adverse effects , Discrimination Learning/drug effects , Female , Fluorouracil/adverse effects , Hippocampus/drug effects , Maze Learning/drug effects , Memory Disorders/psychology , Methotrexate/adverse effects , Neuropsychological Tests , Rats , Rats, Long-Evans
12.
Front Neurosci ; 8: 174, 2014.
Article in English | MEDLINE | ID: mdl-25071426

ABSTRACT

Adult neurogenesis is highly responsive to environmental and physiological factors. The majority of studies to date have examined short-term consequences of enhancing or blocking neurogenesis but long-term changes remain less well understood. Current evidence for age-related declines in neurogenesis warrant further investigation into these long-term changes. In this report we address the hypothesis that early life experience, such as a period of voluntary running in juvenile rats, can alter properties of adult neurogenesis for the remainder of the animal's life. The results indicate that the number of proliferating and differentiating neuronal precursors is not altered in runners beyond the initial weeks post-running, suggesting homeostatic regulation of these processes. However, the rate of neuronal maturation and survival during a 4 week period after cell division was enhanced up to 11 months of age (the end of the study period). This study is the first to show that a transient period of physical activity at a young age promotes changes in neurogenesis that persist over the long-term, which is important for our understanding of the modulation of neurogenesis by exercise with age. Functional integration of adult-born neurons within the hippocampus that resist homeostatic regulation with aging, rather than the absolute number of adult-born neurons, may be an essential feature of adult neurogenesis that promotes the maintenance of neural plasticity in old age.

13.
Neurobiol Aging ; 35(7): 1605-14, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24491422

ABSTRACT

Numerous clinical and epidemiological reports indicate that patients with history of vascular illness such as stroke are more likely to develop dementia as the clinical manifestation of Alzheimer's disease. However, there are little data regarding the pathologic mechanisms that link vascular risk factors to the factors associated with dementia onset. We provide evidence that suggests intriguing detrimental interactions between stroke and ß-amyloid (Aß) toxicity in the hippocampus. Stroke was induced by unilateral striatal injection of endothelin-1, the potent vasoconstrictor. Aß toxicity was modeled by bilateral intracerebroventricular injections of the toxic fragment Aß. Gross morphologic changes in comorbid Aß and stroke rats were enlargement of the lateral ventricles with concomitant shrinkage of the hippocampus. The hippocampus displayed a series of synergistic biochemical alterations, including microgliosis, deposition of Aß precursor protein fragments, and cellular degeneration. In addition, there was bilateral induction of connexin43, reduced neuronal survival, and impaired dendritic development of adult-born immature neurons in the dentate gyrus of these rats compared with either rats alone. Behaviorally, there was impairment in the hippocampal-based discriminative fear-conditioning to context task indicating learning and memory deficit. These results suggest an insight into the relationship between hippocampal atrophy, pathology, and functional impairment. Our work not only highlights the exacerbated pathology that emerges when Aß toxicity and stroke occur comorbidly but also demonstrates that this comorbid rat model exhibits physiopathology that is highly characteristic of the human condition.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Dementia/etiology , Dementia/pathology , Hippocampus/metabolism , Hippocampus/pathology , Stroke/complications , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Protein Precursor/metabolism , Animals , Atrophy , Connexin 43/metabolism , Disease Models, Animal , Endothelin-1 , Humans , Injections, Intraventricular , Male , Stroke/chemically induced , Stroke/pathology , Vasoconstrictor Agents
14.
Psychopharmacology (Berl) ; 231(11): 2311-20, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24343419

ABSTRACT

RATIONALE: Chemotherapy, used for the treatment of cancer, often produces cognitive impairment that has been related to suppression of neurogenesis. Physical exercise, which promotes neurogenesis, is known to improve cognitive function in neurologically challenged animals and humans. It is unknown whether exercise similarly protects against chemotherapy-induced cognitive impairment and whether recovery of neurogenesis is a critical factor. OBJECTIVE: The present study investigated the relationship between hippocampal neurogenesis and cognitive performance in chemotherapy-treated rats that engaged in different amounts of physical activity. METHODS: Groups of rats, housed individually in standard cages or in specially designed cages that allowed unlimited access to a running wheel, received three injections of the chemotherapeutic drugs methotrexate and 5-fluorouracil, or equal volumes of saline. They were then administered the following cognitive tests in a water maze: (1) spatial memory (SM), (2) cued memory, (3) non-matching to sample (NMTS) rule learning; (4) delayed NMTS (DNMTS). Hippocampal neurogenesis was quantified by counting doublecortin-expressing cells in the dentate gyrus. RESULTS: Chemotherapy administered to rats in standard cages resulted in a significant reduction in hippocampal neurogenesis and impaired performance on the SM, NMTS, and DNMTS tasks. In rats receiving chemotherapy and housed in exercise cages, neurogenesis was not suppressed and cognitive performance was similar to controls. CONCLUSIONS: Physical exercise can reduce cognitive deficits that result from chemotherapy and this effect is mediated, at least in part, by preventing suppression of drug-induced hippocampal neurogenesis. The results suggest benefits of exercise in preventing or treating cognitive impairment associated with chemotherapy.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Cognition Disorders/chemically induced , Cognition Disorders/prevention & control , Dentate Gyrus/drug effects , Motor Activity/physiology , Neurogenesis/drug effects , Animals , Body Weight , Cognition Disorders/physiopathology , Cues , Dentate Gyrus/physiopathology , Doublecortin Domain Proteins , Doublecortin Protein , Female , Fluorouracil/adverse effects , Housing, Animal , Immunohistochemistry , Learning/drug effects , Learning/physiology , Memory/drug effects , Memory/physiology , Methotrexate/adverse effects , Microtubule-Associated Proteins/metabolism , Motor Activity/drug effects , Neurogenesis/physiology , Neuropeptides/metabolism , Neuropsychological Tests , Rats, Long-Evans , Spatial Memory/drug effects , Spatial Memory/physiology
15.
Front Neurosci ; 7: 66, 2013.
Article in English | MEDLINE | ID: mdl-23641193

ABSTRACT

Since the remarkable discovery of adult neurogenesis in the mammalian hippocampus, considerable effort has been devoted to unraveling the functional significance of these new neurons. Our group has proposed that a continual turnover of neurons in the DG could contribute to the development of event-unique memory traces that act to reduce interference between highly similar inputs. To test this theory, we implemented a recognition task containing some objects that were repeated across trials as well as some objects that were highly similar, but not identical, to ones previously observed. The similar objects, termed lures, overlap substantially with previously viewed stimuli, and thus, may require hippocampal neurogenesis in order to avoid catastrophic interference. Lifestyle factors such as aerobic exercise and stress have been shown to impact the local neurogenic microenvironment, leading to enhanced and reduced levels of DG neurogenesis, respectively. Accordingly, we hypothesized that healthy young adults who take part in a long-term aerobic exercise regime would demonstrate enhanced performance on the visual pattern separation task, specifically at correctly categorizing lures as "similar." Indeed, those who experienced a proportionally large change in fitness demonstrated a significantly greater improvement in their ability to correctly identify lure stimuli as "similar." Conversely, we expected that those who score high on depression scales, an indicator of chronic stress, would exhibit selective deficits at appropriately categorizing lures. As expected, those who scored high on the Beck Depression Inventory (BDI) were significantly worse than those with relatively lower BDI scores at correctly identifying lures as "similar," while performance on novel and repeated stimuli was identical. Taken together, our results support the hypothesis that adult-born neurons in the DG contribute to the orthogonalization of incoming information.

16.
Ann Neurol ; 74(4): 611-21, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23686887

ABSTRACT

OBJECTIVE: Extrasynaptic γ-aminobutyric acid type A receptors that contain the δ subunit (δGABAA receptors) are highly expressed in the dentate gyrus (DG) subfield of the hippocampus, where they generate a tonic conductance that regulates neuronal activity. GABAA receptor-dependent signaling regulates memory and also facilitates postnatal neurogenesis in the adult DG; however, the role of the δGABAA receptors in these processes is unclear. Accordingly, we sought to determine whether δGABAA receptors regulate memory behaviors, as well as neurogenesis in the DG. METHODS: Memory and neurogenesis were studied in wild-type (WT) mice and transgenic mice that lacked δGABAA receptors (Gabrd(-/-)). To pharmacologically increase δGABAA receptor activity, mice were treated with the δGABAA receptor-preferring agonist 4,5,6,7-tetrahydroisoxazolo(5,4-c)pyridin-3-ol (THIP). Behavioral assays including recognition memory, contextual discrimination, and fear extinction were used. Neurogenesis was studied by measuring the proliferation, survival, migration, maturation, and dendritic complexity of adult-born neurons in the DG. RESULTS: Gabrd(-/-) mice exhibited impaired recognition memory and contextual discrimination relative to WT mice. Fear extinction was also impaired in Gabrd(-/-) mice, although the acquisition of fear memory was enhanced. Neurogenesis was disrupted in Gabrd(-/-) mice as the migration, maturation, and dendritic development of adult-born neurons were impaired. Long-term treatment with THIP facilitated learning and neurogenesis in WT but not Gabrd(-/-) mice. INTERPRETATION: δGABAA receptors promote the performance of certain DG-dependent memory behaviors and facilitate neurogenesis. Furthermore, δGABAA receptors can be pharmacologically targeted to enhance these processes.


Subject(s)
Dentate Gyrus/physiology , Memory/physiology , Neurogenesis/genetics , Receptors, GABA-A/metabolism , Analysis of Variance , Animals , Discrimination, Psychological/physiology , Electroshock/adverse effects , Exploratory Behavior/physiology , GABA Agonists/pharmacology , Isoxazoles/pharmacology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, GABA-A/genetics , Recognition, Psychology/physiology
17.
Autoimmunity ; 46(3): 222-30, 2013 May.
Article in English | MEDLINE | ID: mdl-23249428

ABSTRACT

The essence of autoimmune thyroid disease (AITD) is loss of tolerance of own tissues caused by malfunction of T lymphocytes, which affects the production of antibodies reacting with particular cell structures and tissues. Foxp3(+) regulatory T cells (Tregs) take part in the regulation of immune response and play a leading role in developing immune tolerance through active suppression. The aim of the study was to estimate the expression of CD4+CD25(high), CD4+CD25+CD127(low)FoxP3(+) and CD4+ FoxP3 T cells in patients with Graves' disease (GD) (n = 24, median age 15.5 years), in patients with Hashimoto's thyroiditis (HT) (n = 30, median age 15 years) in comparison with sex- and age-matched healthy control subjects (n = 30, median age 15 years). Polychromatic flow cytometry using a FACSCalibur (BD Biosciences) cytometer was applied to delineate T regulatory cell populations. In untreated patients with Graves' disease and HT we observed a significant decrease in CD4+FoxP3 (p < 0.001, p < 0.01) and CD4+CD25(high) (p < 0.016, p < 0.048) T lymphocytes as compared to the healthy control subjects. After 6-12 months of L-thyroxine therapy in HT cases these phenotypes of Tregs were normalized, yet no such changes were observed during GD therapy. The analysis of CD4+CD25+CD127(low)FoxP3+ T cells in the peripheral blood revealed comparable percentages of these cells in patients with thyroid autoimmune diseases to the controls. We conclude that the reduction number of Tregs with CD4+CD25(high) and CD4+FoxP3 phenotype suggests their role in initiation and development of autoimmune process in thyroid disorders.


Subject(s)
Graves Disease/immunology , Hashimoto Disease/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Thyroxine/therapeutic use , Adolescent , CD4 Antigens/metabolism , Child , Female , Forkhead Transcription Factors/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Male , Thyroid Gland/immunology , Young Adult
18.
Clin Cancer Res ; 18(11): 3112-21, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22467680

ABSTRACT

PURPOSE: Clinical studies indicate that up to 70% of patients with cancer who receive chemotherapy experience cognitive impairment. The present study used a prospective longitudinal design to assess short- and long-term effects of commonly used anticancer drugs on cognitive performance in a mouse model. EXPERIMENTAL DESIGN: Normal mice received three weekly injections of a combination of methotrexate + 5-fluorouracil (CHEMO group) or an equal volume of saline (SAL group). Cognitive tests, measuring different aspects of learning and memory, were administered before treatment, immediately after treatment, and three months later. Structural MRI scanning was conducted at each stage of cognitive testing. RESULTS: The CHEMO group exhibited deficits on cognitive tasks acquired pretreatment [spatial memory, nonmatching-to-sample (NMTS) learning, and delayed NMTS], as well as impaired new learning on two tasks (conditional associative learning, discrimination learning) introduced posttreatment. Consistent with clinical evidence, cognitive deficits were pronounced on tests that are sensitive to hippocampal and frontal lobe dysfunction, but the CHEMO group's poor performance on the discrimination learning problem suggests that impairment is more widespread than previously thought. Cognitive deficits persisted for at least three months after treatment but some recovery was noted, particularly on tests thought to be under frontal lobe control. The MRI tests did not detect brain changes that could be attributed to treatment. CONCLUSIONS: Chemotherapeutic agents can have adverse effects on information acquired pretreatment as well as new learning and memory and, despite some recovery, impairment is long lasting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cognition/drug effects , Animals , Disease Models, Animal , Female , Fluorouracil/adverse effects , Learning/drug effects , Longitudinal Studies , Memory/drug effects , Methotrexate/adverse effects , Mice , Mice, Inbred BALB C , Neuropsychological Tests , Prospective Studies , Time Factors
19.
Behav Brain Res ; 227(2): 464-9, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-21669236

ABSTRACT

Rats, subjected to low-dose irradiation that suppressed hippocampal neurogenesis, or a sham treatment, were administered a visual discrimination task under conditions of high, or low interference. Half of the rats engaged in running activity and the other half did not. In the non-runners, there was no effect of irradiation on learning, or remembering the discrimination response under low interference, but irradiation treatment increased their susceptibility to interference, resulting in loss of memory for the previously learned discrimination. Irradiated rats that engaged in running activity exhibited increased neuronal growth and protection from memory impairment. The results, which show that hippocampal cells generated in adulthood play a role in differentiating between conflicting, context-dependent memories, provide further evidence of the importance of neurogenesis in hippocampus-sensitive memory tasks. The results are consistent with computational models of hippocampal function that specify a central role for neurogenesis in the modulation of interfering influences during learning and memory.


Subject(s)
Discrimination Learning/physiology , Hippocampus/physiology , Memory/physiology , Neurogenesis/physiology , Animals , Male , Neurons/physiology , Physical Conditioning, Animal/physiology , Rats , Rats, Long-Evans
20.
Behav Brain Res ; 227(2): 490-6, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-21893104

ABSTRACT

Our understanding of the hippocampus as a memory-encoding device is greatly helped by our knowledge of neuronal circuits and their plasticity. The trisynaptic hippocampal circuit carrying afferent input from the entorhinal cortex, controlled by a network of inhibitory interneurons and supplemented by modulatory subcortical inputs forms a platform for multiple forms of synaptic plastic mechanisms. Long-term potentiation of synaptic transmission in its various forms is an outstanding example of hippocampal ability to adapt to past neuronal activity. Adult neurogenesis is a profound plastic mechanism incorporating structural and functional changes that were previously thought to be present only in developing neural systems. These powerful forms of plasticity can mask experimental results by compensating for experimentally induced changes in the neurons or circuits. Circuit lesions have been one of the most common techniques in scientific investigations of the hippocampus. Although the effects of such lesions can be quite revealing and ground-breaking, in many cases the results are masked by compensatory mechanisms producing misleading results. This review will highlight such mechanisms and argue that the experimental results, in spite of their shortcomings, can be better understood when viewed in light of our knowledge of the neuronal circuitry, and with guidance by conceptual and computational models. Studies demonstrating a role of neurogenesis in pattern separation and memory interference are a good example of fruitful interaction between modeling and experimental approaches.


Subject(s)
Hippocampus/physiology , Nerve Net/physiology , Neurogenesis/physiology , Neurons/physiology , Animals , Models, Neurological , Neuronal Plasticity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...