Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3836, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714691

ABSTRACT

Exercise has beneficial effects on cognition throughout the lifespan. Here, we demonstrate that specific exercise patterns transform insufficient, subthreshold training into long-term memory in mice. Our findings reveal a potential molecular memory window such that subthreshold training within this window enables long-term memory formation. We performed RNA-seq on dorsal hippocampus and identify genes whose expression correlate with conditions in which exercise enables long-term memory formation. Among these genes we found Acvr1c, a member of the TGF ß family. We find that exercise, in any amount, alleviates epigenetic repression at the Acvr1c promoter during consolidation. Additionally, we find that ACVR1C can bidirectionally regulate synaptic plasticity and long-term memory in mice. Furthermore, Acvr1c expression is impaired in the aging human and mouse brain, as well as in the 5xFAD mouse model, and over-expression of Acvr1c enables learning and facilitates plasticity in mice. These data suggest that promoting ACVR1C may protect against cognitive impairment.


Subject(s)
Activin Receptors, Type I , Epigenesis, Genetic , Hippocampus , Memory, Long-Term , Physical Conditioning, Animal , Animals , Female , Humans , Male , Mice , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Aging/genetics , Aging/physiology , Hippocampus/metabolism , Memory, Long-Term/physiology , Mice, Inbred C57BL , Neuronal Plasticity/genetics , Physical Conditioning, Animal/physiology , Promoter Regions, Genetic
2.
Aging Cell ; 23(6): e14144, 2024 06.
Article in English | MEDLINE | ID: mdl-38500398

ABSTRACT

Aging coincides with the progressive loss of muscle mass and strength, increased adiposity, and diminished physical function. Accordingly, interventions aimed at improving muscle, metabolic, and/or physical health are of interest to mitigate the adverse effects of aging. In this study, we tested a stem cell secretome product, which contains extracellular vesicles and growth, cytoskeletal remodeling, and immunomodulatory factors. We examined the effects of 4 weeks of 2×/week unilateral intramuscular secretome injections (quadriceps) in ambulatory aged male C57BL/6 mice (22-24 months) compared to saline-injected aged-matched controls. Secretome delivery substantially increased whole-body lean mass and decreased fat mass, corresponding to higher myofiber cross-sectional area and smaller adipocyte size, respectively. Secretome-treated mice also had greater whole-body physical function (grip strength and rotarod performance) and had higher energy expenditure and physical activity levels compared to control mice. Furthermore, secretome-treated mice had greater skeletal muscle Pax7+ cell abundance, capillary density, collagen IV turnover, reduced intramuscular lipids, and greater Akt and hormone sensitive lipase phosphorylation in adipose tissue. Finally, secretome treatment in vitro directly enhanced muscle cell growth and IL-6 production, and in adipocytes, it reduced lipid content and improved insulin sensitivity. Moreover, indirect treatment with secretome-treated myotube culture media also enhanced muscle cell growth and adipocyte size reduction. Together, these data suggest that intramuscular treatment with a stem cell secretome improves whole-body metabolism, physical function, and remodels skeletal muscle and adipose tissue in aged mice.


Subject(s)
Adiposity , Aging , Mice, Inbred C57BL , Muscle, Skeletal , Secretome , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Male , Adiposity/drug effects , Mice , Secretome/metabolism , Stem Cells/metabolism
3.
Aging Cell ; 22(9): e13905, 2023 09.
Article in English | MEDLINE | ID: mdl-37334527

ABSTRACT

DNA damage is a central contributor to the aging process. In the brain, a major threat to the DNA is the considerable amount of reactive oxygen species produced, which can inflict oxidative DNA damage. This type of damage is removed by the base excision repair (BER) pathway, an essential DNA repair mechanism, which contributes to genome stability in the brain. Despite the crucial role of the BER pathway, insights into how this pathway is affected by aging in the human brain and the underlying regulatory mechanisms are very limited. By microarray analysis of four cortical brain regions from humans aged 20-99 years (n = 57), we show that the expression of core BER genes is largely downregulated during aging across brain regions. Moreover, we find that expression of many BER genes correlates positively with the expression of the neurotrophin brain-derived neurotrophic factor (BDNF) in the human brain. In line with this, we identify binding sites for the BDNF-activated transcription factor, cyclic-AMP response element-binding protein (CREB), in the promoter of most BER genes and confirm the ability of BDNF to regulate several BER genes by BDNF treatment of mouse primary hippocampal neurons. Together, these findings uncover the transcriptional landscape of BER genes during aging of the brain and suggest BDNF as an important regulator of BER in the human brain.


Subject(s)
Brain-Derived Neurotrophic Factor , DNA Repair , Animals , Humans , Mice , Aging/genetics , Aging/metabolism , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , DNA Repair/genetics , Signal Transduction/genetics
4.
Front Aging ; 3: 796087, 2022.
Article in English | MEDLINE | ID: mdl-35821854

ABSTRACT

Histone modifications are key contributors to the cognitive decline that occurs in aging and Alzheimer's disease. Our lab has previously shown that elevated H3K9me3 in aged mice is correlated with synaptic loss, cognitive impairment and a reduction in brain derived neurotrophic factor (BDNF). However, the mechanism of H3K9me3 regulation remains poorly understood. In this study, we investigated the role of age-associated stressors on H3K9me3 regulation and examined if changes in H3K9me3 were age dependent. We used cultured hippocampal neurons at 6, 12, and 21 days in vitro (DIV) to examine the effect of different stressors on H3K9me3 across neuron ages. We found that the oxidative stressor hydrogen peroxide (H2O2) does not induce H3K9me3 in 12 DIV neurons. Inhibiting BDNF signaling via TrkB-Fc elevated H3K9me3 in 12 and 21 DIV neurons compared to 6 DIV neurons. Antioxidant treatment prevented H3K9me3 elevation in 12 DIV neurons treated with TrkB-Fc and H2O2. H2O2 elevated the epigenetic regulator SIRT1 in 6 DIV neurons but did not increase H3K9me3 levels. Our findings demonstrate that inhibiting BDNF signaling elevates hippocampal H3K9me3 in a manner dependent on in vitro age and oxidative stress.

5.
Front Aging Neurosci ; 13: 798297, 2021.
Article in English | MEDLINE | ID: mdl-34970138

ABSTRACT

Exercise improves cognition in the aging brain and is a key regulator of neuronal plasticity genes such as BDNF. However, the mechanism by which exercise modifies gene expression continues to be explored. The repressive histone modification H3K9me3 has been shown to impair cognition, reduce synaptic density and decrease BDNF in aged but not young mice. Treatment with ETP69, a selective inhibitor of H3K9me3's catalyzing enzyme (SUV39H1), restores synapses, BDNF and cognitive performance. GABA receptor expression, which modulates BDNF secretion, is also modulated by exercise and H3K9me3. In this study, we examined if exercise and ETP69 regulated neuronal plasticity genes by reducing H3K9me3 at their promoter regions. We further determined the effect of age on H3K9me3 promoter binding and neuronal plasticity gene expression. Exercise and ETP69 decreased H3K9me3 at BDNF promoter VI in aged mice, corresponding with an increase in BDNF VI expression with ETP69. Exercise increased GABRA2 in aged mice while increasing BDNF 1 in young mice, and both exercise and ETP69 reduced GABRA2 in young mice. Overall, H3K9me3 repression at BDNF and GABA receptor promoters decreased with age. Our findings suggest that exercise and SUV39H1 inhibition differentially modulate BDNF and GABRA2 expression in an age dependent manner.

6.
Learn Mem ; 26(12): 485-492, 2019 12.
Article in English | MEDLINE | ID: mdl-31732709

ABSTRACT

The beneficial effects of exercise on cognition are well established; however specific exercise parameters regarding the frequency and duration of physical activity that provide optimal cognitive health have not been well defined. Here, we explore the effects of the duration of exercise and sedentary periods on long-term object location memory (OLM) in mice. We use a weak object location training paradigm that is subthreshold for long-term memory formation in sedentary controls, and demonstrate that exercise enables long-term memories to form. We show that 14- and 21-d of running wheel access enables mice to discriminate between familiar and novel object locations after a 24 h delay, while 2- or 7-d running wheel access provides insufficient exercise for such memory enhancement using the subthreshold learning paradigm. After 14- and 21-d of wheel running, exercise-induced cognitive enhancement then decays back to baseline performance following 3-d of sedentary activity. However, exercise-induced cognitive enhancement can be reactivated by an additional period of just 2 d exercise, previously shown to be insufficient to induce cognitive enhancement on its own. The reactivating period of exercise is capable of enhancing memory after three- or seven-sedentary days, but not 14-d. These data suggest a type of "molecular memory" for the exercise stimulus, in that once exercise duration reaches a certain threshold, it establishes a temporal window during which subsequent low-level exercise can capitalize on the neurobiological adaptations induced by the initial period of exercise, enabling it to maintain the benefits on cognitive function. These findings provide new information that may help to guide future clinical studies in exercise.


Subject(s)
Adaptation, Physiological/physiology , Cognition/physiology , Memory, Long-Term/physiology , Physical Conditioning, Animal/physiology , Spatial Memory/physiology , Animals , Behavior, Animal/physiology , Male , Mice , Mice, Inbred C57BL , Time Factors
7.
Neurobiol Aging ; 78: 142-154, 2019 06.
Article in English | MEDLINE | ID: mdl-30927700

ABSTRACT

Exercise has emerged as a powerful variable that can improve cognitive function and delay age-associated cognitive decline and Alzheimer's disease (AD); however, the underlying mechanisms are poorly understood. To determine if protective mechanisms may occur at the transcriptional level, we used microarrays to investigate the relationship between physical activity levels and gene expression patterns in the cognitively intact aged human hippocampus. In parallel, hippocampal gene expression patterns associated with aging and AD were assessed using publicly available microarray data profiling hippocampus from young (20-59 years), cognitively intact aging (73-95 years) and age-matched AD cases. To identify "anti-aging/AD" transcription patterns associated with physical activity, probesets significantly associated with both physical activity and aging/AD were identified and their directions of expression change in each condition were compared. Remarkably, of the 2210 probesets significant in both data sets, nearly 95% showed opposite transcription patterns with physical activity compared with aging/AD. The majority (>70%) of these anti-aging/AD genes showed increased expression with physical activity and decreased expression in aging/AD. Enrichment analysis of the anti-aging/AD genes showing increased expression in association with physical activity revealed strong overrepresentation of mitochondrial energy production and synaptic function, along with axonal function and myelin integrity. Synaptic genes were notably enriched for synaptic vesicle priming, release and recycling, glutamate and GABA signaling, and spine plasticity. Anti-aging/AD genes showing decreased expression in association with physical activity were enriched for transcription-related function (notably negative regulation of transcription). These data reveal that physical activity is associated with a more youthful profile in the hippocampus across multiple biological processes, providing a potential molecular foundation for how physical activity can delay age- and AD-related decline of hippocampal function.


Subject(s)
Aging/genetics , Aging/physiology , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Exercise/physiology , Gene Expression , Hippocampus/metabolism , Hippocampus/physiology , Adult , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Axons/physiology , Cognition , Energy Metabolism/genetics , Humans , Microarray Analysis , Middle Aged , Mitochondria/metabolism , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Synaptic Vesicles/genetics , Synaptic Vesicles/physiology , Young Adult
8.
J Neurosci ; 37(5): 1197-1212, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27986924

ABSTRACT

Long-term potentiation (LTP) is an activity-dependent and persistent increase in synaptic transmission. Currently available techniques to measure LTP are time-intensive and require highly specialized expertise and equipment, and thus are not well suited for screening of multiple candidate treatments, even in animal models. To expand and facilitate the analysis of LTP, here we use a flow cytometry-based method to track chemically induced LTP by detecting surface AMPA receptors in isolated synaptosomes: fluorescence analysis of single-synapse long-term potentiation (FASS-LTP). First, we demonstrate that FASS-LTP is simple, sensitive, and models electrically induced LTP recorded in intact circuitries. Second, we conducted FASS-LTP analysis in two well-characterized Alzheimer's disease (AD) mouse models (3xTg and Tg2576) and, importantly, in cryopreserved human AD brain samples. By profiling hundreds of synaptosomes, our data provide the first direct evidence to support the idea that synapses from AD brain are intrinsically defective in LTP. Third, we used FASS-LTP for drug evaluation in human synaptosomes. Testing a panel of modulators of cAMP and cGMP signaling pathways, FASS-LTP identified vardenafil and Bay-73-6691 (phosphodiesterase-5 and -9 inhibitors, respectively) as potent enhancers of LTP in synaptosomes from AD cases. These results indicate that our approach could provide the basis for protocols to study LTP in both healthy and diseased human brains, a previously unattainable goal. SIGNIFICANCE STATEMENT: Learning and memory depend on the ability of synapses to strengthen in response to activity. Long-term potentiation (LTP) is a rapid and persistent increase in synaptic transmission that is thought to be affected in Alzheimer's disease (AD). However, direct evidence of LTP deficits in human AD brain has been elusive, primarily due to methodological limitations. Here, we analyze LTP in isolated synapses from AD brain using a novel approach that allows testing LTP in cryopreserved brain. Our analysis of hundreds of synapses supports the idea that AD-diseased synapses are intrinsically defective in LTP. Further, we identified pharmacological agents that rescue LTP in AD, thus opening up a new avenue for drug screening and evaluation of strategies for alleviating memory impairments.


Subject(s)
Alzheimer Disease/physiopathology , Long-Term Potentiation/drug effects , Synapses/drug effects , Animals , Cyclic AMP/physiology , Cyclic GMP/physiology , Electric Stimulation , Flow Cytometry , Humans , Male , Mice , Mice, Inbred C57BL , Phosphodiesterase Inhibitors/pharmacology , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Signal Transduction/drug effects , Synaptosomes/drug effects
9.
Alzheimers Dement ; 13(5): 510-519, 2017 May.
Article in English | MEDLINE | ID: mdl-27793643

ABSTRACT

INTRODUCTION: We have comprehensively described the expression profiles of mitochondrial DNA and nuclear DNA genes that encode subunits of the respiratory oxidative phosphorylation (OXPHOS) complexes (I-V) in the hippocampus from young controls, age matched, mild cognitively impaired (MCI), and Alzheimer's disease (AD) subjects. METHODS: Hippocampal tissues from 44 non-AD controls (NC), 10 amnestic MCI, and 18 AD cases were analyzed on Affymetrix Hg-U133 plus 2.0 arrays. RESULTS: The microarray data revealed significant down regulation in OXPHOS genes in AD, particularly those encoded in the nucleus. In contrast, there was up regulation of the same gene(s) in MCI subjects compared to AD and ND cases. No significant differences were observed in mtDNA genes identified in the array between AD, ND, and MCI subjects except one mt-ND6. DISCUSSION: Our findings suggest that restoration of the expression of nuclear-encoded OXPHOS genes in aging could be a viable strategy for blunting AD progression.


Subject(s)
Aging/genetics , Alzheimer Disease/genetics , Cognition Disorders/genetics , Mitochondria/genetics , Oxidative Phosphorylation , Adult , Aged, 80 and over , Autopsy , Female , Hippocampus , Humans , Male , Oligonucleotide Array Sequence Analysis
10.
J Neuroinflammation ; 13(1): 289, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27832807

ABSTRACT

BACKGROUND: While exercise effects on the immune system have received increasing attention in recent years, it remains unclear to what extent gender and fluctuations in sex hormones during menstrual cycle influence immunological responses to exercise. METHODS: We investigated mRNA changes induced through exhaustive exercise (half-marathon; pre-exercise and post-exercise [30 min, 3 h, 24 h] on whole blood cultures ± lipopolysaccharide [LPS] [1 h]) with a specific focus on sex differences (men vs women in luteal phase) as an extension of our previous study. RESULTS: Inflammation related signaling pathways, TLRs, cytosolic DNA sensing and RIG-I like receptors were differentially activated between sexes in LPS-stimulated cultures. Genes differentially regulated between sexes included TNIP-1, TNIP-3, IL-6, HIVEP1, CXCL3, CCR3, IL-8, and CD69, revealing a bias towards less anti-inflammatory gene regulation in women compared to men. In addition, several genes relevant to brain function (KMO, DDIT4, VEGFA, IGF1R, IGF2R, and FGD4) showed differential activation between sexes. Some of these genes (e.g., KMO in women, DDIT4 in both sexes) potentially constitute neuroprotective mechanisms. CONCLUSIONS: These data reveal that the exercise-induced change in gene expression might be gender and menstrual cycle phase dependent.


Subject(s)
Cytokines/metabolism , Endotoxins/pharmacology , Exercise , Gene Expression/drug effects , Leukocytes/drug effects , Leukocytes/metabolism , Signal Transduction/drug effects , Adult , Anthropometry , Athletes , Cells, Cultured , Cytokines/genetics , Female , Gene Expression Profiling , Hormones , Humans , Lipopolysaccharides/pharmacology , Male , Menstrual Cycle/physiology , Sex Factors , Time Factors
11.
Proc Natl Acad Sci U S A ; 112(36): E5078-87, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26305968

ABSTRACT

In the aged brain, synaptic plasticity and memory show increased vulnerability to impairment by the inflammatory cytokine interleukin 1ß (IL-1ß). In this study, we evaluated the possibility that synapses may directly undergo maladaptive changes with age that augment sensitivity to IL-1ß impairment. In hippocampal neuronal cultures, IL-1ß increased the expression of the IL-1 receptor type 1 and the accessory coreceptor AcP (proinflammatory), but not of the AcPb (prosurvival) subunit, a reconfiguration that potentiates the responsiveness of neurons to IL-1ß. To evaluate whether synapses develop a similar heightened sensitivity to IL-1ß with age, we used an assay to track long-term potentiation (LTP) in synaptosomes. We found that IL-1ß impairs LTP directly at the synapse and that sensitivity to IL-1ß is augmented in aged hippocampal synapses. The increased synaptic sensitivity to IL-1ß was due to IL-1 receptor subunit reconfiguration, characterized by a shift in the AcP/AcPb ratio, paralleling our culture data. We suggest that the age-related increase in brain IL-1ß levels drives a shift in IL-1 receptor configuration, thus heightening the sensitivity to IL-1ß. Accordingly, selective blocking of AcP-dependent signaling with Toll-IL-1 receptor domain peptidomimetics prevented IL-1ß-mediated LTP suppression and blocked the memory impairment induced in aged mice by peripheral immune challenge (bacterial lipopolysaccharide). Overall, this study demonstrates that increased AcP signaling, specifically at the synapse, underlies the augmented vulnerability to cognitive impairment by IL-1ß that occurs with age.


Subject(s)
Interleukin-1beta/pharmacology , Neurons/drug effects , Receptors, Interleukin-1 Type I/metabolism , Synapses/metabolism , Age Factors , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/cytology , Hippocampus/metabolism , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/metabolism , Long-Term Potentiation/drug effects , Mice, 129 Strain , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Neurons/metabolism , RNA Interference , Rats, Sprague-Dawley , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/drug effects , Synaptosomes/drug effects , Synaptosomes/metabolism
12.
Neurobiol Aging ; 35(9): 1961-72, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24786631

ABSTRACT

Mild cognitive impairment (MCI) represents a cognitive state intermediate between normal aging and early Alzheimer's disease (AD). To investigate if the molecular signature of MCI parallels the clinical picture, we use microarrays to extensively profile gene expression in 4 cortical brain regions (entorhinal cortex, hippocampus, superior frontal gyrus, post-central gyrus) using the postmortem tissue from cognitively normal aged controls, MCI, and AD cases. Our data reveal that gene expression patterns in MCI are not an extension of aging, and for the most part, are not intermediate between aged controls and AD. Functional enrichment analysis of significant genes revealed prominent upregulation in MCI brains of genes associated with anabolic and biosynthetic pathways (notably transcription, protein biosynthesis, protein trafficking, and turnover) as well as mitochondrial energy generation. In addition, many synaptic genes showed altered expression in MCI, predominantly upregulation, including genes for central components of the vesicle fusion machinery at the synapse, synaptic vesicle trafficking, neurotransmitter receptors, and synaptic structure and stabilization. These data suggest that there is a rebalancing of synaptic transmission in the MCI brain. To investigate if synaptic gene expression levels in MCI were related to cognitive function, Pearson correlation coefficient between the Mini Mental State Examination (MMSE) and region-specific messenger RNA expression were computed for MCI cases. A number of synaptic genes showed strong significant correlations (r > 0.8, p < 0.01) most notably in the entorhinal cortex, with fewer in the hippocampus, and very few in neocortical regions. The synaptic genes with highly significant correlations were predominantly related to synaptic transmission and plasticity, and myelin composition. Unexpectedly, we found that gene expression changes that facilitate synaptic excitability and plasticity were overwhelmingly associated with poorer MMSE, and conversely that gene expression changes that inhibit plasticity were positively associated with MMSE. These data suggest that there are excessive excitability and apparent plasticity in limbic brain regions in MCI, that is associated with impaired synaptic and cognitive function. Such changes would be predicted to contribute to increased excitability, in turn leading to greater metabolic demand and ultimately progressive degeneration and AD, if not controlled.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , Brain/physiopathology , Cognitive Dysfunction/genetics , Gene Expression/genetics , Neuronal Plasticity/genetics , Synaptic Transmission/genetics , Aged , Aged, 80 and over , Aging/genetics , Aging/metabolism , Aging/pathology , Aging/psychology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Brain/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/psychology , Energy Metabolism/genetics , Female , Gene Expression Profiling , Humans , Male , Metabolism/genetics , Microarray Analysis , Mitochondria/genetics , Mitochondria/metabolism , Myelin Sheath/metabolism , Neuropsychological Tests , Protein Biosynthesis/genetics , Protein Transport/genetics , Transcription, Genetic/genetics
13.
Neurobiol Aging ; 35(5): 1002-11, 2014 May.
Article in English | MEDLINE | ID: mdl-24268883

ABSTRACT

Alzheimer's is a crippling neurodegenerative disease that largely affects aged individuals. Decades of research have highlighted age-related changes in calcium homeostasis that occur before and throughout the duration of the disease, and the contributions of such dysregulation to Alzheimer's disease pathogenesis. We report an age-related decrease in expression of the CaV3.1 T-type calcium channel at the level of messenger RNA and protein in both humans and mice that is exacerbated with the presence of Alzheimer's disease. Downregulating T-type calcium channels in N2a cells and the 3xTg-AD mouse model of Alzheimer's disease, by way of pharmacologic inhibition with NNC-55-0396, results in a rapid increase in amyloid beta production via reductions in non-amyloidogenic processing, whereas genetic overexpression of the channel in human embryonic kidney cells expressing amyloid precursor protein produces complementary effects. The age-related decline in CaV3.1 expression may therefore contribute to a pro-amyloidogenic environment in the aging brain and represents a novel opportunity to intervene in the course of Alzheimer's disease pathogenesis.


Subject(s)
Aging/genetics , Brain/metabolism , Calcium Channels, T-Type/metabolism , Calcium/metabolism , Down-Regulation/genetics , Adult , Aged , Aged, 80 and over , Aging/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Humans , Kidney/cytology , Kidney/metabolism , Mice , Middle Aged , Molecular Targeted Therapy , RNA, Messenger/metabolism , Young Adult
14.
J Clin Invest ; 123(10): 4158-69, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23999428

ABSTRACT

Aggregation of tau protein in the brain is associated with a class of neurodegenerative diseases known as tauopathies. FK506 binding protein 51 kDa (FKBP51, encoded by FKBP5) forms a mature chaperone complex with Hsp90 that prevents tau degradation. In this study, we have shown that tau levels are reduced throughout the brains of Fkbp5-/- mice. Recombinant FKBP51 and Hsp90 synergized to block tau clearance through the proteasome, resulting in tau oligomerization. Overexpression of FKBP51 in a tau transgenic mouse model revealed that FKBP51 preserved the species of tau that have been linked to Alzheimer's disease (AD) pathogenesis, blocked amyloid formation, and decreased tangle load in the brain. Alterations in tau turnover and aggregate structure corresponded with enhanced neurotoxicity in mice. In human brains, FKBP51 levels increased relative to age and AD, corresponding with demethylation of the regulatory regions in the FKBP5 gene. We also found that higher FKBP51 levels were associated with AD progression. Our data support a model in which age-associated increases in FKBP51 levels and its interaction with Hsp90 promote neurotoxic tau accumulation. Strategies aimed at attenuating FKBP51 levels or its interaction with Hsp90 have the potential to be therapeutically relevant for AD and other tauopathies.


Subject(s)
Alzheimer Disease/metabolism , HSP90 Heat-Shock Proteins/metabolism , tau Proteins/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid/metabolism , Animals , CA3 Region, Hippocampal/metabolism , Case-Control Studies , DNA Methylation , Female , Gene Expression , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Middle Aged , Proteasome Endopeptidase Complex/metabolism , Protein Multimerization , Protein Structure, Quaternary , Proteolysis , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Young Adult , tau Proteins/chemistry
15.
J Biol Chem ; 288(23): 16937-16948, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23599427

ABSTRACT

We previously found that BDNF-dependent retrograde trafficking is impaired in AD transgenic mouse neurons. Utilizing a novel microfluidic culture chamber, we demonstrate that Aß oligomers compromise BDNF-mediated retrograde transport by impairing endosomal vesicle velocities, resulting in impaired downstream signaling driven by BDNF/TrkB, including ERK5 activation, and CREB-dependent gene regulation. Our data suggest that a key mechanism mediating the deficit involves ubiquitin C-terminal hydrolase L1 (UCH-L1), a deubiquitinating enzyme that functions to regulate cellular ubiquitin. Aß-induced deficits in BDNF trafficking and signaling are mimicked by LDN (an inhibitor of UCH-L1) and can be reversed by increasing cellular UCH-L1 levels, demonstrated here using a transducible TAT-UCH-L1 strategy. Finally, our data reveal that UCH-L1 mRNA levels are decreased in the hippocampi of AD brains. Taken together, our data implicate that UCH-L1 is important for regulating neurotrophin receptor sorting to signaling endosomes and supporting retrograde transport. Further, our results support the idea that in AD, Aß may down-regulate UCH-L1 in the AD brain, which in turn impairs BDNF/TrkB-mediated retrograde signaling, compromising synaptic plasticity and neuronal survival.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Ubiquitin Thiolesterase/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Survival/genetics , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Neuronal Plasticity/genetics , Neurons/metabolism , Neurons/pathology , Protein Transport/genetics , Rats , Receptor, trkB/genetics , Receptor, trkB/metabolism , Signal Transduction/genetics , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin Thiolesterase/genetics
16.
Neuropsychopharmacology ; 38(10): 2027-34, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23615664

ABSTRACT

We demonstrate that exercise enables hippocampal-dependent learning in conditions that are normally subthreshold for encoding and memory formation, and depends on hippocampal induction of brain-derived neurotrophic factor (BDNF) as a key mechanism. Using a weak training paradigm in an object location memory (OLM) task, we show that sedentary mice are unable to discriminate 24 h later between familiar and novel object locations. In contrast, 3 weeks of prior voluntary exercise enables strong discrimination in the spatial memory task. Cognitive benefits of exercise match those attained with post-training sodium butyrate (NaB), a histone deacetylase (HDAC) inhibitor previously shown to enable subthreshold learning. We demonstrate that the enabling effects of exercise and NaB on subthreshold OLM learning are dependent on hippocampal BDNF upregulation, and are blocked by hippocampal infusion of BDNF short-interfering RNA. Exercise and NaB increased bdnf transcripts I and IV, and the increases were associated with BDNF promoter acetylation on H4K8 but not H4K12. These data provide support for the concept that exercise engages epigenetic control mechanisms and serves as a natural stimulus that operates in part like NaB and potentially other HDAC inhibitors, placing the brain into a state of readiness for plasticity.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Butyric Acid/pharmacology , Discrimination Learning/physiology , Hippocampus/metabolism , Memory, Long-Term/physiology , Physical Conditioning, Animal/physiology , Acetylation , Animals , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Discrimination Learning/drug effects , Hippocampus/drug effects , Histone Deacetylase Inhibitors/pharmacology , Male , Memory, Long-Term/drug effects , Mice , Microinjections , Promoter Regions, Genetic , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Up-Regulation
17.
Neurobiol Aging ; 34(6): 1653-61, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23273601

ABSTRACT

Synapses are essential for transmitting, processing, and storing information, all of which decline in aging and Alzheimer's disease (AD). Because synapse loss only partially accounts for the cognitive declines seen in aging and AD, we hypothesized that existing synapses might undergo molecular changes that reduce their functional capacity. Microarrays were used to evaluate expression profiles of 340 synaptic genes in aging (20-99 years) and AD across 4 brain regions from 81 cases. The analysis revealed an unexpectedly large number of significant expression changes in synapse-related genes in aging, with many undergoing progressive downregulation across aging and AD. Functional classification of the genes showing altered expression revealed that multiple aspects of synaptic function are affected, notably synaptic vesicle trafficking and release, neurotransmitter receptors and receptor trafficking, postsynaptic density scaffolding, cell adhesion regulating synaptic stability, and neuromodulatory systems. The widespread declines in synaptic gene expression in normal aging suggests that function of existing synapses might be impaired, and that a common set of synaptic genes are vulnerable to change in aging and AD.


Subject(s)
Aging/genetics , Alzheimer Disease/genetics , Brain Chemistry/genetics , Down-Regulation/genetics , Gene Expression Profiling , Synapses/genetics , Adult , Aged , Aged, 80 and over , Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain Chemistry/physiology , Female , Gene Expression Profiling/methods , Humans , Male , Middle Aged , Synapses/metabolism , Synapses/pathology , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism , Synaptic Vesicles/pathology , Young Adult
18.
J Neuroinflammation ; 9: 264, 2012 Dec 03.
Article in English | MEDLINE | ID: mdl-23206327

ABSTRACT

BACKGROUND: The intricate interactions between the immune, endocrine and central nervous systems shape the innate immune response of the brain. We have previously shown that estradiol suppresses expression of immune genes in the frontal cortex of middle-aged ovariectomized rats, but not in young ones reflecting elevated expression of these genes in middle-aged, ovarian hormone deficient animals. Here, we explored the impact of menopause on the microglia phenotype capitalizing on the differential expression of macrophage-associated genes in quiescent and activated microglia. METHODS: We selected twenty-three genes encoding phagocytic and recognition receptors expressed primarily in microglia, and eleven proinflammatory genes and followed their expression in the rat frontal cortex by real-time PCR. We used young, middle-aged and middle-aged ovariectomized rats to reveal age- and ovariectomy-related alterations. We analyzed the expression of the same set of genes in the postcentral and superior frontal gyrus of pre- and postmenopausal women using raw microarray data from our previous study. RESULTS: Ovariectomy caused up-regulation of four classic microglia reactivity marker genes including Cd11b, Cd18, Cd45 and Cd86. The change was reversible since estradiol attenuated transcriptional activation of the four marker genes. Expression of genes encoding phagocytic and toll-like receptors such as Cd11b, Cd18, C3, Cd32, Msr2 and Tlr4 increased, whereas scavenger receptor Cd36 decreased following ovariectomy. Ovarian hormone deprivation altered the expression of major components of estrogen and neuronal inhibitory signaling which are involved in the control of microglia reactivity. Strikingly similar changes took place in the postcentral and superior frontal gyrus of postmenopausal women. CONCLUSIONS: Based on the overlapping results of rat and human studies we propose that the microglia phenotype shifts from the resting toward the reactive state which can be characterized by up-regulation of CD11b, CD14, CD18, CD45, CD74, CD86, TLR4, down-regulation of CD36 and unchanged CD40 expression. As a result of this shift, microglial cells have lower threshold for subsequent activation in the forebrain of postmenopausal women.


Subject(s)
Aging/metabolism , Antigens, CD/metabolism , Frontal Lobe/metabolism , Gene Expression Regulation/physiology , Menopause/metabolism , Toll-Like Receptor 4/metabolism , Adult , Age Factors , Aged , Animals , Antigens, CD/genetics , Cytokines/genetics , Cytokines/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation/drug effects , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Humans , Menopause/drug effects , Middle Aged , Ovariectomy , Phagocytosis/drug effects , Phagocytosis/physiology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
19.
J Neuroinflammation ; 9: 179, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22824372

ABSTRACT

BACKGROUND: This study undertakes a systematic and comprehensive analysis of brain gene expression profiles of immune/inflammation-related genes in aging and Alzheimer's disease (AD). METHODS: In a well-powered microarray study of young (20 to 59 years), aged (60 to 99 years), and AD (74 to 95 years) cases, gene responses were assessed in the hippocampus, entorhinal cortex, superior frontal gyrus, and post-central gyrus. RESULTS: Several novel concepts emerge. First, immune/inflammation-related genes showed major changes in gene expression over the course of cognitively normal aging, with the extent of gene response far greater in aging than in AD. Of the 759 immune-related probesets interrogated on the microarray, approximately 40% were significantly altered in the SFG, PCG and HC with increasing age, with the majority upregulated (64 to 86%). In contrast, far fewer immune/inflammation genes were significantly changed in the transition to AD (approximately 6% of immune-related probesets), with gene responses primarily restricted to the SFG and HC. Second, relatively few significant changes in immune/inflammation genes were detected in the EC either in aging or AD, although many genes in the EC showed similar trends in responses as in the other brain regions. Third, immune/inflammation genes undergo gender-specific patterns of response in aging and AD, with the most pronounced differences emerging in aging. Finally, there was widespread upregulation of genes reflecting activation of microglia and perivascular macrophages in the aging brain, coupled with a downregulation of select factors (TOLLIP, fractalkine) that when present curtail microglial/macrophage activation. Notably, essentially all pathways of the innate immune system were upregulated in aging, including numerous complement components, genes involved in toll-like receptor signaling and inflammasome signaling, as well as genes coding for immunoglobulin (Fc) receptors and human leukocyte antigens I and II. CONCLUSIONS: Unexpectedly, the extent of innate immune gene upregulation in AD was modest relative to the robust response apparent in the aged brain, consistent with the emerging idea of a critical involvement of inflammation in the earliest stages, perhaps even in the preclinical stage, of AD. Ultimately, our data suggest that an important strategy to maintain cognitive health and resilience involves reducing chronic innate immune activation that should be initiated in late midlife.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , Brain/physiology , Cognition Disorders/immunology , Immunity, Innate , Protein Array Analysis/methods , Adult , Aged , Aged, 80 and over , Aging/genetics , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain/pathology , Cognition Disorders/genetics , Cognition Disorders/pathology , Female , Humans , Male , Middle Aged , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Transcriptome/immunology , Young Adult
20.
Neurobiol Aging ; 33(1): 108-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-20434811

ABSTRACT

The aged canine is a higher animal model that naturally accumulates ß-amyloid (Aß) and shows age-related cognitive decline. However, profiles of Aß accumulation in different species (40 vs. 42), its assembly states, and Aß precursor protein (APP) processing as a function of age remain unexplored. In this study, we show that Aß increases progressively with age as detected in extracellular plaques and biochemically extractable Aß40 and Aß42 species. Soluble oligomeric forms of the peptide, with specific increases in an Aß oligomer migrating at 56 kDa, also increase with age. Changes in APP processing could potentially explain why Aß accumulates, and we show age-related shifts toward decreased total APP protein and nonamyloidogenic (α-secretase) processing coupled with increased amyloidogenic (ß-secretase) cleavage of APP. Importantly, we describe Aß pathology in the cingulate and temporal cortex and provide a description of oligomeric Aß across the canine lifespan. Our findings are in line with observations in the human brain, suggesting that canines are a valuable higher animal model for the study of Aß pathogenesis.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Plaque, Amyloid/metabolism , Aging/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Disease Models, Animal , Dogs , Humans , Plaque, Amyloid/etiology , Temporal Lobe/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...