Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 238: 109651, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37414332

ABSTRACT

Disruption of synaptic function is believed to represent a common pathway contributing to cognitive decline during aging. Optogenetics is a prodigious tool for studying relationships between function and synaptic circuitry but models utilizing viral vectors present limitations. Careful characterization of the functionality of channel rhodopsin in transgenic models is crucial for determining whether they can be used across aging. This includes verifying the light sensitivity of the protein and confirming its ability to generate action potentials in response to light stimulation. We combined in vitro optogenetic methodology and a reduced synaptic preparation of acutely isolated neurons to determine if the ChR2(H134R)-eYFP vGAT mouse model is well-suited for aging studies. We used neurons from young (2-6 mo), middle-aged (10-14 mo) and aged (17-25 mo) bacterial artificial chromosome (BAC) transgenic mouse line with stable expression of the channelrhodopsin-2 (ChR2) variant H134R in GABAergic cell populations. Cellular physiology and calcium dynamics were assessed in basal forebrain (BF) neurons using patch-clamp recording and fura-2 microfluorimetry, alongside 470 nm light stimulation of the transgenic ChR2 channel to characterize a wide array of physiological functions known to decline with age. We found ChR2 expression is functionally maintained across aging, while spontaneous and optically evoked inhibitory postsynaptic currents, and quantal content were decreased. Aged mice also showed an increase in intracellular calcium buffering. These results, which are on par with previous observations, demonstrate that the optogenetic vGAT BAC mouse model is well-suited for investigating age-related changes in calcium signaling and synaptic transmission.


Subject(s)
Optogenetics , Rhodopsin , Mice , Animals , Rhodopsin/genetics , Rhodopsin/metabolism , Optogenetics/methods , Calcium/metabolism , Synaptic Transmission , Mice, Transgenic , Aging , Homeostasis , Channelrhodopsins/genetics , Channelrhodopsins/metabolism
2.
J Neurosci ; 42(6): 1020-1034, 2022 02 09.
Article in English | MEDLINE | ID: mdl-34911797

ABSTRACT

Aging is often associated with cognitive decline and recurrent cellular and molecular impairments. While life-long caloric restriction (CR) may delay age-related cognitive deterioration as well as the onset of neurologic disease, recent studies suggest that late-onset, short-term intermittent fasting (IF), may show comparable beneficial effects as those of life-long CR to improve brain health. We used a new optogenetic aging model to study the effects of late-onset (>18 months), short-term (four to six weeks) IF on age-related changes in GABAergic synaptic transmission, intracellular calcium (Ca2+) buffering, and cognitive status. We used male mice from a bacterial artificial chromosome (BAC) transgenic mouse line with stable expression of the channelrhodopsin-2 (ChR2) variant H134R [VGAT-ChR2(H134R)-EYFP] in a reduced synaptic preparation that allows for specific optogenetic light stimulation on GABAergic synaptic terminals across aging. We performed quantal analysis using the method of failures in this model and show that short-term IF reverses the age-related decrease in quantal content of GABAergic synapses. Likewise, short-term IF also reversed age-related changes in Ca2+ buffering and spontaneous GABAergic synaptic transmission in basal forebrain (BF) neurons of aged mice. Our findings suggest that late-onset short-term IF can reverse age-related physiological impairments in mouse BF neurons but that four weeks IF is not sufficient to reverse age-related cognitive decline.SIGNIFICANCE STATEMENT Here, we demonstrate plasticity of the aging brain and reversal of well-defined hallmarks of brain aging using short-term intermittent fasting (IF) initiated later in life. Few therapeutics are currently available to treat age-related neurologic dysfunction although synaptic dysfunction occurs during aging and neurologic disease is a topic of intense research. Using a new reduced synaptic preparation and optogenetic stimulation we are able to study age-related synaptic mechanisms in greater detail. Several neurophysiological parameters including quantal content were altered during aging and were reversed with short-term IF. These methods can be used to identify potential therapies to reverse physiological dysfunction during aging.


Subject(s)
Aging/pathology , Basal Forebrain/physiology , Calcium/metabolism , Fasting/physiology , Neurons/physiology , Synaptic Transmission/physiology , Aging/physiology , Animals , Basal Forebrain/pathology , Male , Mice , Mice, Transgenic , Neurons/pathology , Optogenetics
3.
Front Aging Neurosci ; 13: 673155, 2021.
Article in English | MEDLINE | ID: mdl-34122049

ABSTRACT

The antidepressant drug amitriptyline is used in the treatment of clinical depression and a variety of neurological conditions such as anxiety, neuropathic pain disorders and migraine. Antidepressants are associated with both therapeutic and untoward effects, and their use in the elderly has tripled since the mid-1990s. Because of this widespread use, we are interested in testing the acute effects of amitriptyline on synaptic transmission at therapeutic concentrations well below those that block voltage-gated calcium channels. We found that 3 µM amitriptyline reduced the frequency of spontaneous GABAergic inhibitory postsynaptic currents (IPSCs) and reduced quantal content in mice at ages of 7-10 mo. and 23-25 mo., suggesting a presynaptic mechanism of action that does not diminish with age. We employed a reduced synaptic preparation of the basal forebrain (BF) and a new optogenetic aging model utilizing a bacterial artificial chromosome (BAC) transgenic mouse line with stable expression of the channelrhodopsin-2 (ChR2) variant H134R specific for GABAergic neurons [VGAT-ChR2(H134R)-EYFP]. This model enables optogenetic light stimulation of specific GABAergic synaptic terminals across aging. Age-related impairment of circadian behavior was used to confirm predictable age-related changes associated with this model. Our results suggest that low concentrations of amitriptyline act presynaptically to reduce neurotransmitter release and that this action is maintained during aging.

4.
Biol Psychiatry ; 86(2): 143-152, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30712809

ABSTRACT

BACKGROUND: Activity-dependent release of brain-derived neurotrophic factor (BDNF) in the medial prefrontal cortex (mPFC) is essential for the rapid and sustained antidepressant actions of ketamine, and a recent study shows a similar requirement for vascular endothelial growth factor (VEGF). Since BDNF is reported to stimulate VEGF expression and/or release in neuroblastoma cells, the present study tested the hypothesis that the actions of BDNF are mediated by VEGF. METHODS: The role of VEGF in the antidepressant behavioral actions of BDNF was tested by intra-mPFC coinfusion of a VEGF neutralizing antibody and by neuron-specific deletion of VEGF. The influence of BDNF on the release of VEGF and the role of VEGF in the neurotrophic actions of BDNF were determined in rat primary cortical neurons. The role of BDNF in the behavioral and neurotrophic actions of VEGF was also determined. RESULTS: The results show that the rapid and sustained antidepressant-like actions of intra-mPFC BDNF are blocked by coinfusion of a VEGF neutralizing antibody, and that neuron-specific mPFC deletion of VEGF blocks the antidepressant-like actions of BDNF. Studies in primary cortical neurons demonstrate that BDNF stimulates the release of VEGF and that BDNF induction of dendrite complexity is blocked by a selective VEGF-fetal liver kinase 1 receptor antagonist. Surprisingly, the results also show reciprocal interactions, indicating that the behavioral and neurotrophic actions of VEGF are dependent on BDNF. CONCLUSIONS: These findings indicate that the antidepressant-like and neurotrophic actions of BDNF require VEGF signaling, but they also demonstrate reciprocal interdependence for BDNF in the actions of VEGF.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/pharmacology , Nootropic Agents/pharmacology , Vascular Endothelial Growth Factor A/physiology , Animals , Antibodies, Blocking/pharmacology , Behavior, Animal/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Microinjections , Neurons/drug effects , Prefrontal Cortex , Primary Cell Culture , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
5.
Am J Psychiatry ; 176(5): 388-400, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30606046

ABSTRACT

OBJECTIVE: The N-methyl-d-aspartate receptor antagonist ketamine produces rapid and sustained antidepressant actions even in patients with treatment-resistant depression. Vascular endothelial growth factor (VEGF) has been implicated in the effects of conventional monoamine-based antidepressants, but the role of VEGF in the rapid antidepressant actions of ketamine remains unclear. The authors examined whether neuronal VEGF signaling in the medial prefrontal cortex (mPFC) mediates the rapid antidepressant actions of ketamine. METHODS: The authors used a combination of approaches, including conditional, neuron-specific knockout of VEGF or its receptor, Flk-1; antibody neutralization; viral-mediated knockdown of Flk-1; and pharmacological inhibitors. Further in vitro and in vivo experiments were performed to examine whether neuronal VEGF signaling was required for the neurotrophic and synaptogenic actions of ketamine that underlie its behavioral actions. RESULTS: The behavioral actions of systemic ketamine are blocked by forebrain excitatory neuron-specific deletion of either VEGF or Flk-1 or by intra-mPFC infusion of a VEGF neutralizing antibody. Moreover, intra-mPFC infusions of VEGF are sufficient to produce rapid ketamine-like behavioral actions, and these effects are blocked by neuron-specific Flk-1 deletion. The results also show that local knockdown of Flk-1 in mPFC excitatory neurons in adulthood blocks the behavioral effects of systemic ketamine. Moreover, inhibition of neuronal VEGF signaling blocks the neurotrophic and synaptogenic effects of ketamine. CONCLUSIONS: Together, these findings indicate that neuronal VEGF-Flk-1 signaling in the mPFC plays an essential role in the antidepressant actions of ketamine.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Neurons/drug effects , Prefrontal Cortex/drug effects , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor Receptor-2/drug effects , Animals , Antibodies, Neutralizing/pharmacology , Behavior, Animal/drug effects , Gene Knockdown Techniques , Gene Knockout Techniques , In Vitro Techniques , Mice , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/metabolism , Quinazolines/pharmacology , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Sci Rep ; 8(1): 13752, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30213954

ABSTRACT

Established fear memory becomes vulnerable to disruption after memory retrieval and extinction; this labile state is critical for inhibiting the return of fear memory. However, the labile state has a very narrow time window after retrieval, and underlying molecular mechanisms are not well known. To that end, we isolated the hippocampus immediately after fear memory retrieval and performed proteomics. We identified Neurobeachin (NBEA), an autism-related regulator of synaptic protein trafficking, to be upregulated after contextual fear memory retrieval. NBEA protein expression was rapid and transient after fear memory retrieval at the synapse. Nbea mRNA was enriched at the synapses, and the rapid induction of NBEA expression was blocked by inhibition of the mammalian target of rapamycin (mTOR)-dependent signaling pathway. Mice with cornu ammonis 1 (CA1)-specific Nbea shRNA knockdown showed normal fear acquisition and contextual fear memory but impaired extinction, suggesting an important role of Nbea in fear memory extinction processes. Consistently, Nbea heterozygotes showed normal fear acquisition and fear memory recall but showed impairment in extinction. Our data suggest that NBEA is necessary either for induction of memory lability or for the physiological process of memory extinction.


Subject(s)
Carrier Proteins/genetics , Fear/physiology , Memory/physiology , Nerve Tissue Proteins/genetics , Animals , Autistic Disorder/genetics , Autistic Disorder/pathology , CA1 Region, Hippocampal/physiology , Carrier Proteins/chemistry , Carrier Proteins/physiology , Chromosome Pairing/genetics , Chromosome Pairing/physiology , Heterozygote , Hippocampus/physiology , Humans , Membrane Proteins , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Protein Transport/genetics , Proteomics , TOR Serine-Threonine Kinases/genetics
7.
Exp Neurobiol ; 27(1): 45-56, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29535569

ABSTRACT

In recent years, as the aging population grows, aging-induced cognitive impairments including dementia and Alzheimer's disease (AD) have become the biggest challenges for global public health and social care. Therefore, the development of potential therapeutic drugs for aging-associated cognitive impairment is essential. Metabolic dysregulation has been considered to be a key factor that affects aging and dementia. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a primary sensor of cellular energy states and regulates cellular energy metabolism. Metformin (1,1-dimethylbiguanide hydrochloride) is a well-known AMPK activator and has been widely prescribed for type 2 diabetes mellitus (T2DM). Since the incidence of T2DM and dementia increases with aging, metformin has been considered to be one of the most promising drugs to target dementia and its related disorders. To that end, here, we tested the efficacy of metformin and HL271, a novel metformin derivative, in aging-induced cognitive decline. Water (control), metformin (100 mg/kg) or HL271 (50 mg/kg) were orally administered to aged mice for two months; then, the mice were subjected to behavioral tests to measure their cognitive function, particularly their contextual, spatial and working memory. AMPK phosphorylation was also measured in the drug-treated mouse brains. Our results show that oral treatment with HL271 (50 mg/kg) but not metformin (100 mg/kg) improved cognitive decline in aged mice. AMPK activation was correlated with behavior recovery after aging-induced cognitive decline. Taken together, these results suggest that the newly synthesized AMPK activator, HL271, could be a potential therapeutic agent to treat age-related cognitive decline.

8.
Biol Psychiatry ; 83(1): 29-37, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28751069

ABSTRACT

BACKGROUND: Brain-derived neurotrophic factor (BDNF) plays a key role in the pathophysiology and treatment of depression. Recent clinical studies demonstrate that scopolamine, a nonselective muscarinic acetylcholine receptor antagonist, produces rapid antidepressant effects in patients with depression. Rodent studies demonstrate that scopolamine increases glutamate transmission and synaptogenesis in the medial prefrontal cortex (mPFC). Here we tested the hypothesis that activity-dependent BDNF release within the mPFC is necessary for the antidepressant actions of scopolamine. METHODS: Behavioral effects of scopolamine were assessed in BDNF Val/Met knock-in mice, in which BDNF processing and release are impaired. In addition, intra-mPFC infusion of a BDNF-neutralizing antibody was performed to test the necessity of BDNF release in driving scopolamine-induced behavioral responses. Further in vivo and in vitro experiments were performed to delineate BDNF-dependent mechanisms underlying the effects of scopolamine. RESULTS: We found that BDNF Met/Met mice have attenuated responses to scopolamine and that anti-BDNF antibody infusions into the mPFC prevented the antidepressant-like behavioral effects of scopolamine. In vitro experiments show that scopolamine rapidly stimulates BDNF release and tropomyosin receptor kinase B-extracellular signal-regulated kinase signaling. Moreover, these effects require alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor activation and are blocked by neuronal silencing. Importantly, pretreatment with verapamil prevented scopolamine-induced behavioral responses and BDNF-tropomyosin receptor kinase B signaling, suggesting that these effects are dependent on activation of voltage-dependent calcium channels. CONCLUSIONS: The results identify an essential role for activity-dependent BDNF release in the rapid antidepressant effects of scopolamine. Attenuation of responses in BDNF Met mice indicates that patients with the Met allele may be less responsive to scopolamine.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Scopolamine/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Calcium Channels, L-Type/metabolism , Cells, Cultured , Depressive Disorder/pathology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice, Transgenic , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats, Sprague-Dawley , Receptor, Muscarinic M1/antagonists & inhibitors , Receptor, Muscarinic M1/metabolism , Receptor, trkB/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Time Factors , Verapamil/pharmacology , gamma-Aminobutyric Acid/metabolism
9.
Neuropsychopharmacology ; 42(6): 1231-1242, 2017 May.
Article in English | MEDLINE | ID: mdl-27634355

ABSTRACT

GLYX-13 is a putative NMDA receptor modulator with glycine-site partial agonist properties that produces rapid antidepressant effects, but without the psychotomimetic side effects of ketamine. Studies were conducted to examine the molecular, cellular, and behavioral actions of GLYX-13 to further characterize the mechanisms underlying the antidepressant actions of this agent. The results demonstrate that a single dose of GLYX-13 rapidly activates the mTORC1 pathway in the prefrontal cortex (PFC), and that infusion of the selective mTORC1 inhibitor rapamycin into the medial PFC (mPFC) blocks the antidepressant behavioral actions of GLYX-13, indicating a requirement for mTORC1 similar to ketamine. The results also demonstrate that GLYX-13 rapidly increases the number and function of spine synapses in the apical dendritic tuft of layer V pyramidal neurons in the mPFC. Notably, GLYX-13 significantly increased the synaptic responses to hypocretin, a measure of thalamocortical synapses, compared with its effects on 5-HT responses, a measure of cortical-cortical responses mediated by the 5-HT2A receptor. Behavioral studies further demonstrate that GLYX-13 does not influence 5-HT2 receptor induced head twitch response or impulsivity in a serial reaction time task (SRTT), whereas ketamine increased responses in both tests. In contrast, both GLYX-13 and ketamine increased attention in the SRTT task, which is linked to hypocretin-thalamocortical responses. The differences in the 5-HT2 receptor synaptic and behavioral responses may be related to the lack of psychotomimetic side effects of GLYX-13 compared with ketamine, whereas regulation of the hypocretin responses may contribute to the therapeutic benefits of both rapid acting antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Ketamine/pharmacology , Oligopeptides/pharmacology , Prefrontal Cortex/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/drug effects , Animals , Antidepressive Agents/administration & dosage , Ketamine/administration & dosage , Male , Mice, Inbred C57BL , Oligopeptides/administration & dosage , Rats , Rats, Sprague-Dawley
10.
Neuropharmacology ; 111: 242-252, 2016 12.
Article in English | MEDLINE | ID: mdl-27634096

ABSTRACT

Recent preclinical and clinical studies demonstrate that three functionally different compounds, the NMDA receptor channel blocker ketamine, mGlu2/3 receptor antagonist LY341495, and NMDA receptor glycine site agent GLYX-13 produce rapid and long lasting antidepressant effects. Furthermore, these agents are reported to stimulate ERK and mTORC1 signaling in brain. Here we used rat primary cortical culture neurons to further examine the cellular actions of these agents. The results demonstrate that low concentrations of all three compounds rapidly increase levels of the phosphorylated and activated forms of ERK and a downstream target of mTORC1, p70S6 kinase, in a concentration and time dependent manner. In addition, each compound rapidly increases BDNF release into the culture media. Further studies demonstrate that induction of BDNF release, as well as stimulation of phospho-ERK is blocked by incubation with an AMPA receptor antagonist. The requirement for AMPA receptor stimulation suggests that the effects of these rapid agents are activity dependent. This possibility is supported by studies demonstrating that neuronal silencing, via incubation with the GABAA receptor agonist muscimol, completely blocks phospho-ERK and BDNF release by each agent. Finally, incubation with each drug for 24 h increases the number and length of neuronal branches. Together, the results demonstrate that these three different rapid acting antidepressant agents increase ERK signaling and BDNF release in an activity dependent manner that leads to increased neuronal complexity. Further studies will be required to determine the exact mechanisms underlying these effects in cultured neurons and in rodent models.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , MAP Kinase Signaling System/drug effects , Amino Acids/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Dendrites/drug effects , GABAergic Neurons/metabolism , Ketamine/pharmacology , Oligopeptides/pharmacology , Primary Cell Culture , Rats , Receptor, trkB/metabolism , Receptors, AMPA/metabolism , Xanthenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...