Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Prog Neurobiol ; 233: 102568, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38216113

ABSTRACT

The Topoisomerase 3B (Top3b) - Tudor domain containing 3 (Tdrd3) protein complex is the only dual-activity topoisomerase complex that can alter both DNA and RNA topology in animals. TOP3B mutations in humans are associated with schizophrenia, autism and cognitive disorders; and Top3b-null mice exhibit several phenotypes observed in animal models of psychiatric and cognitive disorders, including impaired cognitive and emotional behaviors, aberrant neurogenesis and synaptic plasticity, and transcriptional defects. Similarly, human TDRD3 genomic variants have been associated with schizophrenia, verbal short-term memory and educational attainment. However, the importance of Tdrd3 in normal brain function has not been examined in animal models. Here we generated a Tdrd3-null mouse strain and demonstrate that these mice display both shared and unique defects when compared to Top3b-null mice. Shared defects were observed in cognitive behaviors, synaptic plasticity, adult neurogenesis, newborn neuron morphology, and neuronal activity-dependent transcription; whereas defects unique to Tdrd3-deficient mice include hyperactivity, changes in anxiety-like behaviors, olfaction, increased new neuron complexity, and reduced myelination. Interestingly, multiple genes critical for neurodevelopment and cognitive function exhibit reduced levels in mature but not nascent transcripts. We infer that the entire Top3b-Tdrd3 complex is essential for normal brain function, and that defective post-transcriptional regulation could contribute to cognitive and psychiatric disorders.


Subject(s)
Cognitive Dysfunction , Gene Expression Regulation , Animals , Humans , Mice , Amino Acid Sequence , Neurogenesis/genetics , Neuronal Plasticity/genetics , Proteins/genetics , Proteins/metabolism
2.
JACC Clin Electrophysiol ; 9(11): 2219-2235, 2023 11.
Article in English | MEDLINE | ID: mdl-37737772

ABSTRACT

BACKGROUND: The central nervous system's influence on cardiac function is well described; however, direct evidence for signaling from heart to brain remains sparse. Mice with cardiac-selective overexpression of adenylyl cyclase type 8 (TGAC8) display elevated heart rate/contractility and altered neuroautonomic surveillance. OBJECTIVES: In this study the authors tested whether elevated adenylyl cyclase type 8-dependent signaling at the cardiac cell level affects brain activity and behavior. METHODS: A telemetry system was used to record electrocardiogram (ECG) and electroencephalogram (EEG) in TGAC8 and wild-type mice simultaneously. The Granger causality statistical approach evaluated variations in the ECG/EEG relationship. Mouse behavior was assessed via elevated plus maze, open field, light-dark box, and fear conditioning tests. Transcriptomic and proteomic analyses were performed on brain tissue lysates. RESULTS: Behavioral testing revealed increased locomotor activity in TGAC8 that included a greater total distance traveled (+43%; P < 0.01), a higher average speed (+38%; P < 0.01), and a reduced freezing time (-45%; P < 0.01). Dual-lead telemetry recording confirmed a persistent heart rate elevation with a corresponding reduction in ECG-R-waves interval variability and revealed increased EEG-gamma activity in TGAC8 vs wild-type. Bioinformatic assessment of hippocampal tissue indicated upregulation of dopamine 5, gamma-aminobutyric acid A, and metabotropic glutamate 1/5 receptors, major players in gamma activity generation. Granger causality analyses of ECG and EEG recordings showed a marked increase in informational flow between the TGAC8 heart and brain. CONCLUSIONS: Perturbed signals arising from the heart cause changes in brain activity, altering mouse behavior. More specifically, the brain interprets augmented myocardial humoral/functional output as a "sustained exercise-like" situation and responds by activating central nervous system output controlling locomotion.


Subject(s)
Adenylyl Cyclases , Behavior , Heart , Proteomics , Animals , Mice , Adenylyl Cyclases/metabolism , Brain/metabolism , Heart/physiology , Behavior/physiology
3.
Glia ; 71(10): 2473-2494, 2023 10.
Article in English | MEDLINE | ID: mdl-37401784

ABSTRACT

Nogo-A, B, and C are well described members of the reticulon family of proteins, most well known for their negative regulatory effects on central nervous system (CNS) neurite outgrowth and repair following injury. Recent research indicates a relationship between Nogo-proteins and inflammation. Microglia, the brain's immune cells and inflammation-competent compartment, express Nogo protein, although specific roles of the Nogo in these cells is understudied. To examine inflammation-related effects of Nogo, we generated a microglial-specific inducible Nogo KO (MinoKO) mouse and challenged the mouse with a controlled cortical impact (CCI) traumatic brain injury (TBI). Histological analysis shows no difference in brain lesion sizes between MinoKO-CCI and Control-CCI mice, although MinoKO-CCI mice do not exhibit the levels of ipsilateral lateral ventricle enlargement as injury matched controls. Microglial Nogo-KO results in decreased lateral ventricle enlargement, microglial and astrocyte immunoreactivity, and increased microglial morphological complexity compared to injury matched controls, suggesting decreased tissue inflammation. Behaviorally, healthy MinoKO mice do not differ from control mice, but automated tracking of movement around the home cage and stereotypic behavior, such as grooming and eating (termed cage "activation"), following CCI is significantly elevated. Asymmetrical motor function, a deficit typical of unilaterally brain lesioned rodents, was not detected in CCI injured MinoKO mice, while the phenomenon was present in CCI injured controls 1-week post-injury. Overall, our studies show microglial Nogo as a negative regulator of recovery following brain injury. To date, this is the first evaluation of the roles microglial specific Nogo in a rodent injury model.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Nogo Proteins , Animals , Mice , Brain Injuries/pathology , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Inflammation/metabolism , Mice, Inbred C57BL , Microglia/metabolism , Nogo Proteins/metabolism
4.
Aging Cell ; 22(4): e13793, 2023 04.
Article in English | MEDLINE | ID: mdl-36846960

ABSTRACT

Olfactory dysfunction is a prevalent symptom and an early marker of age-related neurodegenerative diseases in humans, including Alzheimer's and Parkinson's Diseases. However, as olfactory dysfunction is also a common symptom of normal aging, it is important to identify associated behavioral and mechanistic changes that underlie olfactory dysfunction in nonpathological aging. In the present study, we systematically investigated age-related behavioral changes in four specific domains of olfaction and the molecular basis in C57BL/6J mice. Our results showed that selective loss of odor discrimination was the earliest smelling behavioral change with aging, followed by a decline in odor sensitivity and detection while odor habituation remained in old mice. Compared to behavioral changes related with cognitive and motor functions, smelling loss was among the earliest biomarkers of aging. During aging, metabolites related with oxidative stress, osmolytes, and infection became dysregulated in the olfactory bulb, and G protein coupled receptor-related signaling was significantly down regulated in olfactory bulbs of aged mice. Poly ADP-ribosylation levels, protein expression of DNA damage markers, and inflammation increased significantly in the olfactory bulb of older mice. Lower NAD+ levels were also detected. Supplementation of NAD+ through NR in water improved longevity and partially enhanced olfaction in aged mice. Our studies provide mechanistic and biological insights into the olfaction decline during aging and highlight the role of NAD+ for preserving smelling function and general health.


Subject(s)
Olfaction Disorders , Smell , Humans , Mice , Animals , Olfaction Disorders/diagnosis , Olfaction Disorders/pathology , Mice, Inbred C57BL , NAD/metabolism , Aging/pathology , DNA Damage , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Inflammation/metabolism
5.
Cell Rep ; 41(8): 111682, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36417863

ABSTRACT

In vitro studies suggest that mapping the spatiotemporal complexity of nuclear factor κB (NF-κB) signaling is essential to understanding its function. The lack of tools to directly monitor NF-κB proteins in vivo has hindered such efforts. Here, we introduce reporter mice with the endogenous RelA (p65) or c-Rel labeled with distinct fluorescent proteins and a double knockin with both subunits labeled. Overcoming hurdles in simultaneous live-cell imaging of RelA and c-Rel, we show that quantitative features of signaling reflect the identity of activating ligands, differ between primary and immortalized cells, and shift toward c-Rel in microglia from aged brains. RelA:c-Rel heterodimer is unexpectedly depleted in the nuclei of stimulated cells. Trajectories of subunit co-expression in immune lineages reveal a reduction at key cell maturation stages. These results demonstrate the power of these reporters in gaining deeper insights into NF-κB biology, with the spectral complementarity of the labeled NF-κB proteins enabling diverse applications.


Subject(s)
NF-kappa B , Signal Transduction , Mice , Animals , NF-kappa B/metabolism , Cell Nucleus/metabolism , Aging , Cell Line
6.
PLoS Genet ; 18(11): e1010506, 2022 11.
Article in English | MEDLINE | ID: mdl-36441670

ABSTRACT

Short telomeres induce a DNA damage response (DDR) that evokes apoptosis and senescence in human cells. An extant question is the contribution of telomere dysfunction-induced DDR to the phenotypes observed in aging and telomere biology disorders. One candidate is RAP1, a telomere-associated protein that also controls transcription at extratelomeric regions. To distinguish these roles, we generated a knockin mouse carrying a mutated Rap1, which was incapable of binding telomeres and did not result in eroded telomeres or a DDR. Primary Rap1 knockin embryonic fibroblasts showed decreased RAP1 expression and re-localization away from telomeres, with an increased cytosolic distribution akin to that observed in human fibroblasts undergoing telomere erosion. Rap1 knockin mice were viable, but exhibited transcriptomic alterations, proinflammatory cytokine/chemokine signaling, reduced lifespan, and decreased healthspan with increased body weight/fasting blood glucose levels, spontaneous tumor incidence, and behavioral deficits. Taken together, our data present mechanisms distinct from telomere-induced DDR that underlie age-related phenotypes.


Subject(s)
Shelterin Complex , Telomere , Animals , Humans , Mice , Longevity , Phenotype , Telomere/genetics , Telomere Shortening
7.
Mol Neurodegener ; 17(1): 60, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36064424

ABSTRACT

BACKGROUND: Although ɑ-synuclein (ɑ-syn) spreading in age-related neurodegenerative diseases such as Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) has been extensively investigated, the role of aging in the manifestation of disease remains unclear. METHODS: We explored the role of aging and inflammation in the pathogenesis of synucleinopathies in a mouse model of DLB/PD initiated by intrastriatal injection of ɑ-syn preformed fibrils (pff). RESULTS: We found that aged mice showed more extensive accumulation of ɑ-syn in selected brain regions and behavioral deficits that were associated with greater infiltration of T cells and microgliosis. Microglial inflammatory gene expression induced by ɑ-syn-pff injection in young mice had hallmarks of aged microglia, indicating that enhanced age-associated pathologies may result from inflammatory synergy between aging and the effects of ɑ-syn aggregation. Based on the transcriptomics analysis projected from Ingenuity Pathway Analysis, we found a network that included colony stimulating factor 2 (CSF2), LPS related genes, TNFɑ and poly rl:rC-RNA as common regulators. CONCLUSIONS: We propose that aging related inflammation (eg: CSF2) influences outcomes of pathological spreading of ɑ-syn and suggest that targeting neuro-immune responses might be important in developing treatments for DLB/PD.


Subject(s)
Parkinson Disease , Synucleinopathies , Animals , Brain/metabolism , Disease Models, Animal , Inflammation/metabolism , Mice , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
8.
Sci Adv ; 8(35): eabo1440, 2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36054363

ABSTRACT

Physical exercise is rewarding and protective against drug abuse and addiction. However, the neural mechanisms underlying these actions remain unclear. Here, we report that long-term wheel-running produced a more robust increase in c-fos expression in the red nucleus (RN) than in other brain regions. Anatomic and functional assays demonstrated that most RN magnocellular portion (RNm) neurons are glutamatergic. Wheel-running activates a subset of RNm glutamate neurons that project to ventral tegmental area (VTA) dopamine neurons. Optogenetic stimulation of this pathway was rewarding, as assessed by intracranial self-stimulation and conditioned place preference, whereas optical inhibition blocked wheel-running behavior. Running wheel access decreased cocaine self-administration and cocaine seeking during extinction. Last, optogenetic stimulation of the RNm-to-VTA glutamate pathway inhibited responding to cocaine. Together, these findings indicate that physical exercise activates a specific RNm-to-VTA glutamatergic pathway, producing exercise reward and reducing cocaine intake.

9.
Elife ; 112022 05 04.
Article in English | MEDLINE | ID: mdl-35507394

ABSTRACT

Many age-associated changes in the human hematopoietic system have been reproduced in murine models; however, such changes have not been as robustly explored in rats despite the fact these larger rodents are more physiologically similar to humans. We examined peripheral blood of male F344 rats ranging from 3 to 27 months of age and found significant age-associated changes with distinct leukocyte population shifts. We report CD25+ CD4+ population frequency is a strong predictor of healthy aging, generate a model using blood parameters, and find rats with blood profiles that diverge from chronologic age indicate debility; thus, assessments of blood composition may be useful for non-lethal disease profiling or as a surrogate measure for efficacy of aging interventions. Importantly, blood parameters and DNA methylation alterations, defined distinct juncture points during aging, supporting a non-linear aging process. Our results suggest these inflection points are important considerations for aging interventions. Overall, we present rat blood aging metrics that can serve as a resource to evaluate health and the effects of interventions in a model system physiologically more reflective of humans.


Our blood contains many types of white blood cells, which play important roles in defending the body against infections and other threats to our health. The number of these cells changes with age, and this in turn contributes to many other alterations that happen in the body as we get older. For example, the immune system generally gets weaker at fighting infections and preventing other cells from developing into cancer. On top of that, the white blood cells themselves can become cancerous, resulting in several types of blood cancer that are more likely to happen in older people. Many previous studies have examined how the number of white blood cells changes with age in humans and mice. However, our understanding of this process in rats is still poor, despite the fact that the way the human body works has more in common with the rat body than the mouse body. Here, Yanai, Dunn et al. have studied samples of blood from rats between three to 27 months old. The experiments found that it is possible to accurately predict the age of healthy rats by measuring the frequency of populations of white blood cells, especially a certain type known as CD25+ CD4+ cells. If the animals had any form of illness, their predicted age deviated from their actual age. Furthermore, while some changes in the blood were gradual and continuous, others displayed distinct shifts when the rats reached specific ages. In the future, these findings may be used as a tool to help researchers diagnose illnesses in rats before the animals develop symptoms, or to more easily establish if a treatment is having a positive effect on the rats' health. The work of Yanai, Dunn et al. also provides new insights into aging that could potentially aid the design of new screening methods to predict cancer and intervene using a model system that is more similar to humans.


Subject(s)
Aging , Leukocytes , Aging/genetics , Animals , DNA Methylation , Male , Mice , Population Dynamics , Rats , Rats, Inbred F344
10.
Alzheimers Dement ; 18(11): 2327-2340, 2022 11.
Article in English | MEDLINE | ID: mdl-35234334

ABSTRACT

OBJECTIVE: Evaluating the efficacy of 3,6'-dithioPomalidomide in 5xFAD Alzheimer's disease (AD) mice to test the hypothesis that neuroinflammation is directly involved in the development of synaptic/neuronal loss and cognitive decline. BACKGROUND: Amyloid-ß (Aß) or tau-focused clinical trials have proved unsuccessful in mitigating AD-associated cognitive impairment. Identification of new drug targets is needed. Neuroinflammation is a therapeutic target in neurodegenerative disorders, and TNF-α a pivotal neuroinflammatory driver. NEW HYPOTHESIS: AD-associated chronic neuroinflammation directly drives progressive synaptic/neuronal loss and cognitive decline. Pharmacologically mitigating microglial/astrocyte activation without altering Aß generation will define the role of neuroinflammation in AD progression. MAJOR CHALLENGES: Difficulty of TNF-α-lowering compounds reaching brain, and identification of a therapeutic-time window to preserve the beneficial role of neuroinflammatory processes. LINKAGE TO OTHER MAJOR THEORIES: Microglia/astroglia are heavily implicated in maintenance of synaptic plasticity/function in healthy brain and are disrupted by Aß. Mitigation of chronic gliosis can restore synaptic homeostasis/cognitive function.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Animals , Mice , Amyloid beta-Peptides , Cognition , Disease Models, Animal , Mice, Inbred C57BL , Mice, Transgenic , Microglia , Neuroinflammatory Diseases , Neuronal Plasticity , Tumor Necrosis Factor-alpha
11.
J Gerontol A Biol Sci Med Sci ; 77(5): 956-963, 2022 05 05.
Article in English | MEDLINE | ID: mdl-34718551

ABSTRACT

Age-dependent differences in methylation at specific cytosine-guanine (CpG) sites have been used in "epigenetic clock" formulas to predict age. Deviations of epigenetic age from chronological age are informative of health status and are associated with adverse health outcomes, including mortality. In most cases, epigenetic clocks are performed on methylation from DNA extracted from circulating blood cells. However, the effect of neoplastic cells in the circulation on estimation and interpretation of epigenetic clocks is not well understood. Here, we explored this using Fischer 344 (F344) rats, a strain that often develops large granular lymphocyte leukemia (LGLL). We found clear histological markers of LGLL pathology in the spleens and livers of 27 out of 61 rats aged 17-27 months. We assessed DNA methylation by reduced representation bisulfite sequencing with coverage of 3 million cytosine residues. Although LGLL broadly increased DNA methylation variability, it did not change epigenetic aging. Despite this, the inclusion of rats with LGLL in clock training sets significantly altered predictor selection probability at 83 of 121 commonly utilized CpG sites. Furthermore, models trained on rat samples that included individuals with LGLL had greater absolute age error than those trained exclusively rats free of LGLL (39% increase; p < .0001). We conclude that the epigenetic signals for aging and LGLL are distinct, such that LGLL assessment is not necessary for valid measures of epigenetic age in F344 rats. The precision and architecture of constructed epigenetic clock formulas, however, can be influenced by the presence of neoplastic hematopoietic cells in training set populations.


Subject(s)
DNA Methylation , Leukemia, Large Granular Lymphocytic , Aging/genetics , Animals , Cytosine , Epigenesis, Genetic , Leukemia, Large Granular Lymphocytic/genetics , Rats , Rats, Inbred F344
12.
Pharmacol Biochem Behav ; 205: 173186, 2021 06.
Article in English | MEDLINE | ID: mdl-33836219

ABSTRACT

Serotonin is widely implicated as a modulator of brain reward function. However, laboratory studies have not yielded a consensus on which specific reward-related processes are influenced by serotonin and in what manner. Here we explored the role of serotonin in cue-reward learning in mice. In a first series of experiments, we found that acute administration of the serotonin reuptake inhibitors citalopram, fluoxetine, or duloxetine all reduced lever pressing reinforced on an FR1 schedule with presentation of a cue that had been previously paired with delivery of food. However, citalopram had no effect on responding that was reinforced with both cue and food on an FR1 schedule. Furthermore, citalopram did not affect nose poke responses that produced no auditory, visual, or proprioceptive cues but were reinforced with food pellets on a progressive ratio schedule. We next performed region-specific knock out of tryptophan hydroxylase-2 (Tph2), the rate-limiting enzyme in serotonin synthesis. Viral delivery of Cre recombinase was targeted to dorsal or median raphe nuclei (DRN, MRN), the major sources of ascending serotonergic projections. MRN but not DRN knockouts were impaired in development of cue-elicited approach during Pavlovian conditioning; both groups were subsequently hyper-responsive when lever pressing for cue presentation. The inhibitory effect of citalopram was attenuated in DRN but not MRN knockouts. Our findings are in agreement with prior studies showing serotonin to suppress responding for conditioned reinforcers. Furthermore, these results suggest an inhibitory role of MRN serotonin neurons in the initial attribution of motivational properties to a reward-predictive cue, but not in its subsequent maintenance. In contrast, the DRN appears to promote the reduction of motivational value attached to a cue when it is presented repeatedly in the absence of primary reward.


Subject(s)
Conditioning, Classical/drug effects , Dorsal Raphe Nucleus/metabolism , Motivation/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/metabolism , Animals , Citalopram/pharmacology , Cues , Duloxetine Hydrochloride/pharmacology , Female , Fluoxetine/pharmacology , Gene Knockout Techniques/methods , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Reward , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
13.
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
14.
Pharmacol Biochem Behav ; 202: 173104, 2021 03.
Article in English | MEDLINE | ID: mdl-33444596

ABSTRACT

Methamphetamine (METH) is a highly addictive psychostimulant. The continuous use of METH may lead to its abuse and neurotoxicity that have been associated with METH-induced increases in release of dopamine (DA) and glutamate in the brain. METH action in DA has been shown to be mediated by redistribution of DA from vesicles into cytoplasm via vesicular monoamine transporter 2 (VMAT2) and the subsequent reversal of membrane DA transporter (DAT), while little is known about the mechanisms underlying METH-induced glutamate release. Recent studies indicate that a subpopulation of midbrain DA neurons co-expresses VMAT2 and vesicular glutamate transporter 2 (VGLUT2). Therefore, we hypothesized that METH-induced glutamate release may in part originate from such a dual phenotype of DA neurons. To test this hypothesis, we used Cre-LoxP techniques to selectively delete VGLUT2 from midbrain DA neurons, and then examined nucleus accumbens (NAc) DA and glutamate responses to METH using in vivo brain microdialysis between DA-VGLUT2-KO mice and their VGLUT2-HET littermates. We found that selective deletion of VGLUT2 from DA neurons did not significantly alter basal levels of extracellular DA and glutamate, but attenuated METH-induced increases in extracellular levels of DA and glutamate. In addition, DA-VGLUT2-KO mice also displayed lower locomotor response to METH than VGLUT2-HET control mice. These findings, for the first time, suggest that cell-type specific VGLUT2 expression in DA neurons plays an important role in the behavioral and neurochemical effects of METH. Glutamate corelease from DA neurons may in part contributes to METH-induced increase in NAc glutamate release.


Subject(s)
Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Gene Deletion , Glutamic Acid/metabolism , Mesencephalon/metabolism , Methamphetamine/pharmacology , Signal Transduction/drug effects , Vesicular Glutamate Transport Protein 2/genetics , Amphetamine-Related Disorders/metabolism , Animals , Behavior, Animal/drug effects , Gene Knockout Techniques , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis/methods , Nucleus Accumbens/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
15.
Elife ; 92020 11 12.
Article in English | MEDLINE | ID: mdl-33179594

ABSTRACT

Robust biomarkers of aging have been developed from DNA methylation in humans and more recently, in mice. This study aimed to generate a novel epigenetic clock in rats-a model with unique physical, physiological, and biochemical advantages-by incorporating behavioral data, unsupervised machine learning, and network analysis to identify epigenetic signals that not only track with age, but also relates to phenotypic aging. Reduced representation bisulfite sequencing (RRBS) data was used to train an epigenetic age (DNAmAge) measure in Fischer 344 CDF (F344) rats. This measure correlated with age at (r = 0.93) in an independent sample, and related to physical functioning (p=5.9e-3), after adjusting for age and cell counts. DNAmAge was also found to correlate with age in male C57BL/6 mice (r = 0.79), and was decreased in response to caloric restriction. Our signatures driven by CpGs in intergenic regions that showed substantial overlap with H3K9me3, H3K27me3, and E2F1 transcriptional factor binding.


Subject(s)
Aging/metabolism , Biological Clocks/physiology , Epigenesis, Genetic/physiology , Heterochromatin/metabolism , Aging/genetics , Aging/physiology , Animals , Biological Clocks/genetics , Biomarkers , DNA Methylation/genetics , DNA Methylation/physiology , Male , Mice, Inbred C57BL , Phenotype , Rats , Rats, Inbred F344 , Unsupervised Machine Learning
16.
Nat Commun ; 11(1): 3143, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561719

ABSTRACT

Topoisomerase 3ß (Top3ß) is the only dual-activity topoisomerase in animals that can change topology for both DNA and RNA, and facilitate transcription on DNA and translation on mRNAs. Top3ß mutations have been linked to schizophrenia, autism, epilepsy, and cognitive impairment. Here we show that Top3ß knockout mice exhibit behavioural phenotypes related to psychiatric disorders and cognitive impairment. The mice also display impairments in hippocampal neurogenesis and synaptic plasticity. Notably, the brains of the mutant mice exhibit impaired global neuronal activity-dependent transcription in response to fear conditioning stress, and the affected genes include many with known neuronal functions. Our data suggest that Top3ß is essential for normal brain function, and that defective neuronal activity-dependent transcription may be a mechanism by which Top3ß deletion causes cognitive impairment and psychiatric disorders.


Subject(s)
Cognitive Dysfunction/genetics , DNA Topoisomerases, Type I/genetics , Mental Disorders/genetics , Neurogenesis/genetics , Neuronal Plasticity/genetics , Animals , Behavior Observation Techniques , Behavior, Animal , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/pathology , Disease Models, Animal , Female , Hippocampus/cytology , Hippocampus/diagnostic imaging , Hippocampus/growth & development , Hippocampus/pathology , Humans , Magnetic Resonance Imaging , Male , Mental Disorders/diagnosis , Mental Disorders/pathology , Mice , Mice, Knockout , Neurons/pathology , Stereotaxic Techniques , Synaptic Potentials/genetics , Transcription, Genetic/physiology
17.
Proc Natl Acad Sci U S A ; 117(15): 8611-8615, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32229573

ABSTRACT

Electrical or optogenetic stimulation of lateral hypothalamic (LH) GABA neurons induces rapid vigorous eating in sated animals. The dopamine system has been implicated in the regulation of feeding. Previous work has suggested that a subset of LH GABA neurons projects to the ventral tegmental area (VTA) and targets GABA neurons, inhibiting them and thereby disinhibiting dopaminergic activity and release. Furthermore, stimulation-induced eating is attenuated by dopamine lesions or receptor antagonists. Here we explored the involvement of dopamine in LH stimulation-induced eating. LH stimulation caused sated mice to pick up pellets of standard chow with latencies that varied based on stimulation intensity; once food was picked up, animals ate for the remainder of the 60-s stimulation period. However, lesion of VTA GABA neurons failed to disrupt this effect. Moreover, direct stimulation of VTA or substantia nigra dopamine cell bodies failed to induce food approach or eating. Looking further, we found that some LH GABA fibers pass through the VTA to more caudal sites, where they synapse onto neurons near the locus coeruleus (LC). Similar eating was induced by stimulation of LH GABA terminals or GABA cell bodies in this peri-LC region. Lesion of peri-LC GABA neurons blocked LH stimulation-induced eating, establishing them as a critical downstream circuit element for LH neurons. Surprisingly, lesions did not alter body weight, suggesting that this system is not involved in the hunger or satiety mechanisms that govern normal feeding. Thus, we present a characterization of brain circuitry that may promote overeating and contribute to obesity.


Subject(s)
Dopaminergic Neurons/metabolism , Eating/physiology , Feeding Behavior/physiology , GABAergic Neurons/metabolism , Hypothalamic Area, Lateral/physiology , Ventral Tegmental Area/physiology , Animals , Behavior, Animal , Dopamine/metabolism , Dopaminergic Neurons/cytology , Female , GABAergic Neurons/cytology , Hypothalamic Area, Lateral/cytology , Male , Mice , Neural Pathways , Receptors, GABA-A/metabolism , Reward , Ventral Tegmental Area/cytology , gamma-Aminobutyric Acid/metabolism
18.
Commun Biol ; 3(1): 139, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32198461

ABSTRACT

Findings have shown that anterior insular cortex (aIC) lesions disrupt the maintenance of drug addiction, while imaging studies suggest that connections between amygdala and aIC participate in drug-seeking. However, the role of the BLA â†’ aIC pathway in rewarding contextual memory has not been assessed. Using a cre-recombinase under the tyrosine hydroxylase (TH+) promoter mouse model to induce a real-time conditioned place preference (rtCPP), we show that photoactivation of TH+ neurons induced electrophysiological responses in VTA neurons, dopamine release and neuronal modulation in the aIC. Conversely, memory retrieval induced a strong release of glutamate, dopamine, and norepinephrine in the aIC. Only intra-aIC blockade of the glutamatergic N-methyl-D-aspartate receptor accelerated rtCPP extinction. Finally, photoinhibition of glutamatergic BLA → aIC pathway produced disinhibition of local circuits in the aIC, accelerating rtCPP extinction and impairing reinstatement. Thus, activity of the glutamatergic projection from the BLA to the aIC is critical for maintenance of rewarding contextual memory.


Subject(s)
Basolateral Nuclear Complex/metabolism , Behavior, Animal , Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Memory , Neural Pathways/metabolism , Reward , Adrenergic Neurons/metabolism , Animals , Basolateral Nuclear Complex/cytology , Cerebral Cortex/cytology , Conditioning, Psychological , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Extinction, Psychological , Female , Integrases/genetics , Integrases/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition , Neural Pathways/cytology , Norepinephrine/metabolism , Tyrosine 3-Monooxygenase/genetics
19.
Proc Natl Acad Sci U S A ; 115(49): E11532-E11541, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30442663

ABSTRACT

A subset of midbrain dopamine (DA) neurons express vesicular glutamate transporter 2 (VgluT2), which facilitates synaptic vesicle loading of glutamate. Recent studies indicate that such expression can modulate DA-dependent reward behaviors, but little is known about functional consequences of DA neuron VgluT2 expression in neurodegenerative diseases like Parkinson's disease (PD). Here, we report that selective deletion of VgluT2 in DA neurons in conditional VgluT2-KO (VgluT2-cKO) mice abolished glutamate release from DA neurons, reduced their expression of brain-derived neurotrophic factor (BDNF) and tyrosine receptor kinase B (TrkB), and exacerbated the pathological effects of exposure to the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Furthermore, viral rescue of VgluT2 expression in DA neurons of VglutT2-cKO mice restored BDNF/TrkB expression and attenuated MPTP-induced DA neuron loss and locomotor impairment. Together, these findings indicate that VgluT2 expression in DA neurons is neuroprotective. Genetic or environmental factors causing reduced expression or function of VgluT2 in DA neurons may place some individuals at increased risk for DA neuron degeneration. Therefore, maintaining physiological expression and function of VgluT2 in DA neurons may represent a valid molecular target for the development of preventive therapeutic interventions for PD.


Subject(s)
Dopaminergic Neurons/physiology , Vesicular Glutamate Transport Protein 2/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Electrophysiological Phenomena , Gene Expression Regulation , Glutamic Acid/metabolism , MPTP Poisoning , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Motor Activity/drug effects , Motor Activity/genetics , Mutation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Vesicular Glutamate Transport Protein 2/genetics
20.
Neuropsychopharmacology ; 43(4): 680-689, 2018 03.
Article in English | MEDLINE | ID: mdl-28984293

ABSTRACT

Brain stimulation has identified two central subsets of stimulation sites with motivational relevance. First, there is a large and disperse set of sites where stimulation is reinforcing, increasing the frequency of the responses it follows, and second, a much more restricted set of sites where-along with reinforcement-stimulation also has drive-like effects, instigating feeding, copulation, predation, and other motivated acts in otherwise sated or peaceful animals. From this work a dispersed but synaptically interconnected network of reinforcement circuitry is emerging: it includes afferents to the ventral tegmental area and substantia nigra; the dopamine systems themselves; glutamatergic afferents to the striatum; and one of two dopamine-receptor-expressing efferent pathways of the striatum. Stimulation of a limited subset of these sites, including descending inhibitory medial forebrain bundle fibers, induces both feeding and reinforcement, and suggests the possibility of a subset of fibers where stimulation has both drive-like and reinforcing effects. This review stresses the common findings of sites and connectivity between electrical and optogenetic studies of core drive and reinforcement sites. By doing so, it suggests the biological importance of optogenetic follow-up of less-publicized electrical stimulation findings. Such studies promise not only information about origins, neurotransmitters, and connectivity of related networks, by covering more sensory and at least one putative motor component they also promote a much deeper understanding of the breadth of motivational function.


Subject(s)
Brain/physiology , Motivation/physiology , Nerve Net/physiology , Neurotransmitter Agents/physiology , Reinforcement, Psychology , Reward , Animals , Corpus Striatum/physiology , Humans , Neural Pathways/physiology , Substantia Nigra/physiology , Ventral Tegmental Area/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...