Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Sci Rep ; 14(1): 15944, 2024 07 10.
Article in English | MEDLINE | ID: mdl-38987437

ABSTRACT

Assessment of spatial learning abilities is central to behavioral neuroscience and a useful tool for animal model validation and drug development. However, biases introduced by the apparatus, environment, or experimentalist represent a critical challenge to the test validity. We have recently developed the Modified Barnes Maze (MBM) task, a spatial learning paradigm that overcomes inherent behavioral biases of animals in the classical Barnes maze. The specific combination of spatial strategies employed by mice is often considered representative of the level of cognitive resources used. Herein, we have developed a convolutional neural network-based classifier of exploration strategies in the MBM that can effectively provide researchers with enhanced insights into cognitive traits in mice. Following validation, we compared the learning performance of female and male C57BL/6J mice, as well as that of Ts65Dn mice, a model of Down syndrome, and 5xFAD mice, a model of Alzheimer's disease. Male mice exhibited more effective navigation abilities than female mice, reflected in higher utilization of effective spatial search strategies. Compared to wildtype controls, Ts65Dn mice exhibited delayed usage of spatial strategies despite similar success rates in completing this spatial task. 5xFAD mice showed increased usage of non-spatial strategies such as Circling that corresponded to higher latency to reach the target and lower success rate. These data exemplify the need for deeper strategy classification tools in dissecting complex cognitive traits. In sum, we provide a machine-learning-based strategy classifier that extends our understanding of mice's spatial learning capabilities while enabling a more accurate cognitive assessment.


Subject(s)
Maze Learning , Mice, Inbred C57BL , Neural Networks, Computer , Spatial Learning , Animals , Male , Female , Mice , Spatial Learning/physiology , Disease Models, Animal , Alzheimer Disease , Behavior, Animal
2.
Glia ; 72(6): 1117-1135, 2024 06.
Article in English | MEDLINE | ID: mdl-38450767

ABSTRACT

Williams syndrome (WS) is a genetic neurodevelopmental disorder caused by a heterozygous microdeletion, characterized by hypersociability and unique neurocognitive abnormalities. Of the deleted genes, GTF2I has been linked to hypersociability in WS. We have recently shown that Gtf2i deletion from forebrain excitatory neurons, referred to as Gtf2i conditional knockout (cKO) mice leads to multi-faceted myelination deficits associated with the social behaviors affected in WS. These deficits were potentially mediated also by microglia, as they present a close relationship with oligodendrocytes. To study the impact of altered myelination, we characterized these mice in terms of microglia over the course of development. In postnatal day 30 (P30) Gtf2i cKO mice, cortical microglia displayed a more ramified state, as compared with wild type (controls). However, postnatal day 4 (P4) microglia exhibited high proliferation rates and an elevated activation state, demonstrating altered properties related to activation and inflammation in Gtf2i cKO mice compared with control. Intriguingly, P4 Gtf2i cKO-derived microglial cells exhibited significantly elevated myelin phagocytosis in vitro compared to control mice. Lastly, systemic injection of clemastine to P4 Gtf2i cKO and control mice until P30, led to a significant interaction between genotypes and treatments on the expression levels of the phagocytic marker CD68, and a significant reduction of the macrophage/microglial marker Iba1 transcript levels in the cortex of the Gtf2i cKO treated mice. Our data thus implicate microglia as important players in WS, and that early postnatal manipulation of microglia might be beneficial in treating inflammatory and myelin-related pathologies.


Subject(s)
Transcription Factors, TFIII , Transcription Factors, TFII , Williams Syndrome , Mice , Animals , Microglia , Williams Syndrome/genetics , Neurons/metabolism , Disease Models, Animal , Transcription Factors, TFIII/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
3.
Front Aging Neurosci ; 15: 1067196, 2023.
Article in English | MEDLINE | ID: mdl-36819726

ABSTRACT

Introduction: Recently, interest has emerged in subjective cognitive decline (SCD) as a potential precursor to Alzheimer's disease (AD) dementia. Whether individuals with SCD harbor brain alterations in midlife, when AD-related pathology begins, is yet to be elucidated. Furthermore, the role of apolipoprotein ε4 (APOE ε4) allele, a robust AD risk factor, in the relationship between SCD and brain alterations is unknown. We examined whether APOE genotype modulates the association of SCD with brain measures in individuals at high AD risk. Methods: Middle-aged adults with parental history of AD dementia underwent magnetic resonance imaging (MRI) and the Memory Functioning Questionnaire. Regression analysis tested the extent to which SCD was associated with activation during an functional MRI (fMRI) working-memory task, and white-matter microstructure. APOE ε4 genotype was tested as a moderator. Results: Among APOE ε4 carriers, but not among non-carriers, SCD was associated with higher activation in the anterior cingulate (p = 0.003), inferior, middle, and superior frontal cortices (p = 0.041, p = 0.048, p = 0.037, respectively); and with lower fractional anisotropy in the uncinate fasciculus (p = 0.002), adjusting for age, sex, and education. Conclusion: In middle aged, cognitively normal individuals at high AD risk, higher SCD was associated with greater brain alterations possibly reflecting incipient AD pathology. When accompanied by a family history of AD and an APOE ε4 allele, SCD may have important clinical value, allowing a window for early intervention and for participants' stratification in AD prevention clinical trials.

4.
Proc Natl Acad Sci U S A ; 119(49): e2210766119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36442093

ABSTRACT

Transient soluble oligomers of amyloid-ß (Aß) are toxic and accumulate early prior to insoluble plaque formation and cognitive impairment in Alzheimer's disease (AD). Synthetic cyclic D,L-α-peptides (e.g., 1) self-assemble into cross ß-sheet nanotubes, react with early Aß species (1-3 mers), and inhibit Aß aggregation and toxicity in stoichiometric concentrations, in vitro. Employing a semicarbazide as an aza-glycine residue with an extra hydrogen-bond donor to tune nanotube assembly and amyloid engagement, [azaGly6]-1 inhibited Aß aggregation and toxicity at substoichiometric concentrations. High-resolution NMR studies revealed dynamic interactions between [azaGly6]-1 and Aß42 residues F19 and F20, which are pivotal for early dimerization and aggregation. In an AD mouse model, brain positron emission tomography (PET) imaging using stable 64Cu-labeled (aza)peptide tracers gave unprecedented early amyloid detection in 44-d presymptomatic animals. No tracer accumulation was detected in the cortex and hippocampus of 44-d-old 5xFAD mice; instead, intense PET signal was observed in the thalamus, from where Aß oligomers may spread to other brain parts with disease progression. Compared with standard 11C-labeled Pittsburgh compound-B (11C-PIB), which binds specifically fibrillar Aß plaques, 64Cu-labeled (aza)peptide gave superior contrast and uptake in young mouse brain correlating with Aß oligomer levels. Effectively crossing the blood-brain barrier (BBB), peptide 1 and [azaGly6]-1 reduced Aß oligomer levels, prolonged lifespan of AD transgenic Caenorhabditis elegans, and abated memory and behavioral deficits in nematode and murine AD models. Cyclic (aza)peptides offer novel promise for early AD diagnosis and therapy.


Subject(s)
Alzheimer Disease , Amyloidosis , Animals , Mice , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Early Diagnosis , Amyloid beta-Peptides , Plaque, Amyloid , Amyloidogenic Proteins
6.
Front Neurosci ; 16: 937663, 2022.
Article in English | MEDLINE | ID: mdl-36033613

ABSTRACT

Obesity and hyperglycemia are risk factors for cognitive decline and for the development of Alzheimer's Disease (AD). Bariatric surgery is an effective treatment for obesity that was shown to improve cognitive decline in obese patients. Bariatric surgery was shown to exert weight loss independent effects on metabolic diseases such as type 2 diabetes. We tested whether sleeve gastrectomy (SG), a common bariatric surgery, can affect the cognitive impairment in lean, normoglycemic female 5xFAD mice, a genetic model for AD. 5xFAD mice and wild-type (WT) littermates underwent SG or sham surgery at the age of 5 months and were tested for metabolic, behavioral, and molecular phenotypes 90 days later. SG led to a reduction in blood glucose levels and total plasma cholesterol levels in 5xFAD mice without inducing weight loss. However, the surgery did not affect the outcomes of long-term spatial memory tests in these mice. Analysis of ß-Amyloid plaques corroborated the behavioral studies in showing no effect of surgery on the molecular phenotype of 5xFAD mice. In conclusion, SG leads to an improved metabolic profile in lean female 5xFAD mice without inducing weight loss but does not affect the brain pathology or behavioral phenotype. Our results suggest that the positive effects of bariatric surgery on cognitive decline in obese patients are likely attributed to weight loss and improvement in obesity sequelae, and not to weight loss independent effects of surgery.

7.
Neuromolecular Med ; 24(4): 399-404, 2022 12.
Article in English | MEDLINE | ID: mdl-35411485

ABSTRACT

Microglia, the primary brain-resident immune cells, protect the brain from various harmful pathogens, insulting and maintaining its homeostasis by phagocytosing extracellular particles. How microglia are metabolically regulated by their microenvironment remains largely elusive. Here, we investigated how extracellular lactate, which is abundant in the brain and dynamically changes in pathological states, affects microglial phagocytotic ability. We show that L-lactate reduces microglia phagocytic capacity in a Hydroxycarboxylic Acid Receptor 1 but not Monocarboxylate transporter 1-dependent manner. Our findings point to a potential role for extracellular lactate in suppressing the phagocytic activity of microglial cells in homeostasis and inflammatory conditions.


Subject(s)
Lactic Acid , Microglia , Phagocytosis , Receptors, G-Protein-Coupled , Signal Transduction
8.
Vaccines (Basel) ; 10(4)2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35455279

ABSTRACT

Individuals with Down syndrome (DS) are among the groups with the highest risk for severe COVID-19. Better understanding of the efficacy and risks of COVID-19 vaccines for individuals with DS may help improve uptake of vaccination. The T21RS COVID-19 Initiative launched an international survey to obtain information on safety and efficacy of COVID-19 vaccines for individuals with DS. De-identified survey data collected between March and December 2021 were analyzed. Of 2172 individuals with DS, 1973 (91%) had received at least one vaccine dose (57% BNT162b2), 107 (5%) were unvaccinated by choice, and 92 (4%) were unvaccinated for other reasons. Most participants had either no side effects (54%) or mild ones such as pain at the injection site (29%), fatigue (12%), and fever (7%). Severe side effects occurred in <0.5% of participants. About 1% of the vaccinated individuals with DS contracted COVID-19 after vaccination, and all recovered. Individuals with DS who were unvaccinated by choice were more likely to be younger, previously recovered from COVID-19, and also unvaccinated against other recommended vaccines. COVID-19 vaccines have been shown to be safe for individuals with DS and effective in terms of resulting in minimal breakthrough infections and milder disease outcomes among fully vaccinated individuals with DS.

9.
Geroscience ; 44(4): 2171-2194, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35357643

ABSTRACT

Intermittent fasting (IF) remains the most effective intervention to achieve robust anti-aging effects and attenuation of age-related diseases in various species. Epigenetic modifications mediate the biological effects of several environmental factors on gene expression; however, no information is available on the effects of IF on the epigenome. Here, we first found that IF for 3 months caused modulation of H3K9 trimethylation (H3K9me3) in the cerebellum, which in turn orchestrated a plethora of transcriptomic changes involved in robust metabolic switching processes commonly observed during IF. Second, a portion of both the epigenomic and transcriptomic modulations induced by IF was remarkably preserved for at least 3 months post-IF refeeding, indicating that memory of IF-induced epigenetic changes was maintained. Notably, though, we found that termination of IF resulted in a loss of H3K9me3 regulation of the transcriptome. Collectively, our study characterizes the novel effects of IF on the epigenetic-transcriptomic axis, which controls myriad metabolic processes. The comprehensive analyses undertaken in this study reveal a molecular framework for understanding how IF impacts the metabolo-epigenetic axis of the brain and will serve as a valuable resource for future research.


Subject(s)
Epigenomics , Transcriptome , Fasting , Gene Expression Profiling , Brain
10.
Pharmacol Ther ; 235: 108122, 2022 07.
Article in English | MEDLINE | ID: mdl-35114285

ABSTRACT

Amyloid precursor protein (APP) is an evolutionarily conserved transmembrane protein and a well-characterized precursor protein of amyloid-beta (Aß) peptides, which accumulate in the brains of individuals with Alzheimer's disease (AD)-related pathologies. Aß has been extensively investigated since the amyloid hypothesis in AD was proposed. Besides Aß, previous studies on APP and its proteolytic cleavage products have suggested their diverse pathological and physiological functions. However, their roles still have not been thoroughly understood. In this review, we extensively discuss the evolutionarily-conserved biology of APP, including its structure and processing pathway, as well as recent findings on the physiological roles of APP and its fragments in the central nervous system and peripheral nervous system. We have also elaborated upon the current status of APP-targeted therapeutic approaches for AD treatment by discussing inhibitors of several proteases participating in APP processing, including α-, ß-, and γ-secretases. Finally, we have highlighted the future perspectives pertaining to further research and the potential clinical role of APP.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Humans
12.
Vaccine ; 39(34): 4817-4829, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34294479

ABSTRACT

Aging-related decline in immune functions, termed immunosenescence, is a primary cause of reduced protective responses to vaccines in the elderly, due to impaired induction of cellular and humoral responses to new antigens (Ag), especially if the response is T cell dependent. The result is a more severe morbidity following infections, more prolonged and frequent hospitalization, and a higher mortality rate than in the general population. Therefore, there is an increasing need to develop vaccination strategies that overcome immunosenescence, especially for aging-related diseases such as Alzheimer's disease (AD). Here we report a new vaccination strategy harnessing memory-based immunity, which is less affected by aging. We found that aged C57BL/6 and 5xFAD mice exhibit a dramatic reduction in anti-Amyloid-ß (Aß) antibody (Ab) production. We aimed to reverse this process by inducing memory response at a young age. To this end, young mice were primed with the vaccine carrier Hepatitis B surface antigen (HBsAg). At an advanced age, these mice were immunized with an Aß1-11 fused to HBsAg. This vaccination scheme elicited a markedly higher Aß-specific antibody titer than vaccinating aged unprimed mice with the same construct. Importantly, this vaccine strategy more efficiently reduced cerebral Aß levels and altered microglial phenotype. Overall, we provide evidence that priming with an exogenous Ag carrier can overcome impaired humoral responses to self-antigens in the elderly, paving the route for a potent immunotherapy to AD.


Subject(s)
Alzheimer Disease , Peptide Fragments , Aged , Alzheimer Disease/prevention & control , Amyloid beta-Peptides , Animals , Disease Models, Animal , Hepatitis B Vaccines , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
Front Immunol ; 12: 621440, 2021.
Article in English | MEDLINE | ID: mdl-34248930

ABSTRACT

The risk of severe outcomes following respiratory tract infections is significantly increased in individuals over 60 years, especially in those with chronic medical conditions, i.e., hypertension, diabetes, cardiovascular disease, dementia, chronic respiratory disease, and cancer. Down Syndrome (DS), the most prevalent intellectual disability, is caused by trisomy-21 in ~1:750 live births worldwide. Over the past few decades, a substantial body of evidence has accumulated, pointing at the occurrence of alterations, impairments, and subsequently dysfunction of the various components of the immune system in individuals with DS. This associates with increased vulnerability to respiratory tract infections in this population, such as the influenza virus, respiratory syncytial virus, SARS-CoV-2 (COVID-19), and bacterial pneumonias. To emphasize this link, here we comprehensively review the immunobiology of DS and its contribution to higher susceptibility to severe illness and mortality from respiratory tract infections.


Subject(s)
Down Syndrome/immunology , Immune System/physiology , Orthomyxoviridae/physiology , Respiratory Syncytial Viruses/physiology , Respiratory Tract Infections/immunology , SARS-CoV-2/physiology , Virus Diseases/immunology , Adult , Animals , COVID-19 , Down Syndrome/genetics , Down Syndrome/mortality , Humans , Pneumonia , Respiratory Tract Infections/genetics , Respiratory Tract Infections/mortality , Risk , Virus Diseases/genetics , Virus Diseases/mortality
15.
Neuromolecular Med ; 23(4): 445-448, 2021 12.
Article in English | MEDLINE | ID: mdl-33871752

ABSTRACT

Adult hippocampal neurogenesis is a dynamic process involved in cognitive functions, like learning and memory. Numerous intrinsic and extrinsic factors regulate and affect hippocampal neurogenesis. An exceptionally beneficial external factor is physical exercise due to the impact of the lactate accumulated during physical effort on neural plasticity. Lactate has recently emerged as one of the most interesting and potent factors in health and disease due to its involvement in the metabolism and signaling of most, if not all, of the cells in the CNS. Herein, we illustrate the effects induced by lactate on the different cell types within the neurogenic niche, in light of their described roles in regulating adult hippocampal neurogenesis.


Subject(s)
Lactic Acid , Neurogenesis , Adult , Cognition/physiology , Hippocampus , Humans , Lactic Acid/pharmacology , Neuronal Plasticity
16.
Nat Commun ; 12(1): 2185, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846335

ABSTRACT

The function of B cells in Alzheimer's disease (AD) is not fully understood. While immunoglobulins that target amyloid beta (Aß) may interfere with plaque formation and hence progression of the disease, B cells may contribute beyond merely producing immunoglobulins. Here we show that AD is associated with accumulation of activated B cells in circulation, and with infiltration of B cells into the brain parenchyma, resulting in immunoglobulin deposits around Aß plaques. Using three different murine transgenic models, we provide counterintuitive evidence that the AD progression requires B cells. Despite expression of the AD-fostering transgenes, the loss of B cells alone is sufficient to reduce Aß plaque burden and disease-associated microglia. It reverses behavioral and memory deficits and restores TGFß+ microglia, respectively. Moreover, therapeutic depletion of B cells at the onset of the disease retards AD progression in mice, suggesting that targeting B cells may also benefit AD patients.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , B-Lymphocytes/immunology , Disease Progression , Lymphocyte Depletion , Amyloid beta-Peptides/metabolism , Animals , Female , Hippocampus/pathology , Humans , Interleukin-1beta/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Phenotype , Plaque, Amyloid/metabolism , Transforming Growth Factor beta/metabolism
17.
Commun Biol ; 4(1): 329, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712740

ABSTRACT

Maternal antibodies (MAbs) protect against infections in immunologically-immature neonates. Maternally transferred immunity may also be harnessed to target diseases associated with endogenous protein misfolding and aggregation, such as Alzheimer's disease (AD) and AD-pathology in Down syndrome (DS). While familial early-onset AD (fEOAD) is associated with autosomal dominant mutations in the APP, PSEN1,2 genes, promoting cerebral Amyloid-ß (Aß) deposition, DS features a life-long overexpression of the APP and DYRK1A genes, leading to a cognitive decline mediated by Aß overproduction and tau hyperphosphorylation. Although no prenatal screening for fEOAD-related mutations is in clinical practice, DS can be diagnosed in utero. We hypothesized that anti-Aß MAbs might promote the removal of early Aß accumulation in the central nervous system of human APP-expressing mice. To this end, a DNA-vaccine expressing Aß1-11 was delivered to wild-type female mice, followed by mating with 5xFAD males, which exhibit early Aß plaque formation. MAbs reduce the offspring's cortical Aß levels 4 months after antibodies were undetectable, along with alleviating short-term memory deficits. MAbs elicit a long-term shift in microglial phenotype in a mechanism involving activation of the FcγR1/Syk/Cofilin pathway. These data suggest that maternal immunization can alleviate cognitive decline mediated by early Aß deposition, as occurs in EOAD and DS.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/prevention & control , Alzheimer Vaccines/administration & dosage , Amyloid beta-Peptides/metabolism , Antibodies/metabolism , Brain/enzymology , Peptide Fragments/administration & dosage , Phagocytosis , Receptors, IgG/metabolism , Syk Kinase/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Antibodies/immunology , Behavior, Animal , Brain/immunology , Brain/pathology , Cognition , Disease Models, Animal , Female , Immunization , Male , Memory , Mice, Inbred C57BL , Mice, Transgenic , Microglia/enzymology , Microglia/immunology , Microglia/pathology , Peptide Fragments/immunology , Phenotype , Plaque, Amyloid , Signal Transduction , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
18.
Neuromolecular Med ; 23(4): 561-571, 2021 12.
Article in English | MEDLINE | ID: mdl-33660221

ABSTRACT

The current SARS-CoV-2 outbreak, which causes COVID-19, is particularly devastating for individuals with chronic medical conditions, in particular those with Down Syndrome (DS) who often exhibit a higher prevalence of respiratory tract infections, immune dysregulation and potential complications. The incidence of Alzheimer's disease (AD) is much higher in DS than in the general population, possibly increasing further the risk of COVID-19 infection and its complications. Here we provide a biological overview with regard to specific susceptibility of individuals with DS to SARS-CoV-2 infection as well as data from a recent survey on the prevalence of COVID-19 among them. We see an urgent need to protect people with DS, especially those with AD, from COVID-19 and future pandemics and focus on developing protective measures, which also include interventions by health systems worldwide for reducing the negative social effects of long-term isolation and increased periods of hospitalization.


Subject(s)
COVID-19/epidemiology , COVID-19/virology , Disease Susceptibility , Down Syndrome/epidemiology , Adolescent , Adult , Alzheimer Disease/complications , Alzheimer Disease/epidemiology , Alzheimer Disease/immunology , COVID-19/complications , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Comorbidity , Disease Susceptibility/immunology , Disease Susceptibility/virology , Down Syndrome/complications , Down Syndrome/immunology , Female , Hospitalization , Humans , Immune System/abnormalities , Incidence , Male , Pandemics/prevention & control , Prevalence , Risk Factors , Vaccination/methods
19.
Neuromolecular Med ; 23(3): 335-338, 2021 09.
Article in English | MEDLINE | ID: mdl-33386577

ABSTRACT

High-intensity interval training (HIIT) and strength exercise are known to improve health markers, such as cardiovascular health, metabolic health, and cognitive function, as well as to reduce all-cause mortality. High-Intensity Functional Training (HIFT) is a training paradigm derived from both HIIT and strength exercise to elicit greater muscle recruitment than repetitive aerobic exercises, thereby improving both cardiovascular fitness and strength parameters. Herein, we provide a focused review of the known molecular mechanisms that underlie the beneficial effects of HIFT on cardiovascular, metabolic, and cognitive functions.


Subject(s)
High-Intensity Interval Training , Resistance Training , Animals , Cardiovascular Physiological Phenomena , Cognition/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Mice , Muscle Proteins/metabolism , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/physiology , Neurogenesis/physiology , Organ Size , Physical Conditioning, Animal/physiology , Running/physiology
20.
Front Behav Neurosci ; 14: 584052, 2020.
Article in English | MEDLINE | ID: mdl-33281575

ABSTRACT

It is well-established that physical exercise in humans improves cognitive functions, such as executive functions, pattern separation, and working memory. It is yet unknown, however, whether spatial learning, long known to be affected by exercise in rodents, is also affected in humans. In order to address this question, we recruited 20 healthy young male adults (18-30 years old) divided into exercise and control groups (n = 10 in each group). The exercise group performed three sessions per week of mild-intensity aerobic exercise for 12 weeks, while the control group was instructed not to engage in any physical activity. Both groups performed maximal oxygen uptake (VO2max) tests to assess their cardiovascular fitness at baseline and every 4 weeks through the 12 weeks of the training program. The effects of mild aerobic exercise were tested on performance in two different virtual reality (VR)-based spatial learning tasks: (1) virtual Morris water maze (VMWM) and (2) virtual Radial arm water maze (VRAWM). Subjects were tested in both tasks at baseline prior to the training program and at the end of 12 weeks training program. While the mild-intensity aerobic exercise did not affect subjects' VO2max parameters, mean time to anaerobic threshold increased for the exercise group compared with control. No effect was observed, however, on performance in the VMWM or VRAWM between the two groups. Based on these results, we suggest that mild-intensity aerobic exercise does not improve spatial learning and memory in young, healthy adults.

SELECTION OF CITATIONS
SEARCH DETAIL