Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Proc Natl Acad Sci U S A ; 120(45): e2301534120, 2023 Nov 07.
Article in English | MEDLINE | ID: mdl-37903257

ABSTRACT

L-type voltage-gated calcium (Ca2+) channels (L-VGCC) dysfunction is implicated in several neurological and psychiatric diseases. While a popular therapeutic target, it is unknown whether molecular mechanisms leading to disrupted L-VGCC across neurodegenerative disorders are conserved. Importantly, L-VGCC integrate synaptic signals to facilitate a plethora of cellular mechanisms; however, mechanisms that regulate L-VGCC channel density and subcellular compartmentalization are understudied. Herein, we report that in disease models with overactive mammalian target of rapamycin complex 1 (mTORC1) signaling (or mTORopathies), deficits in dendritic L-VGCC activity are associated with increased expression of the RNA-binding protein (RBP) Parkinsonism-associated deglycase (DJ-1). DJ-1 binds the mRNA coding for the alpha and auxiliary Ca2+ channel subunits CaV1.2 and α2δ2, and represses their mRNA translation, only in the disease states, specifically preclinical models of tuberous sclerosis complex (TSC) and Alzheimer's disease (AD). In agreement, DJ-1-mediated repression of CaV1.2/α2δ2 protein synthesis in dendrites is exaggerated in mouse models of AD and TSC, resulting in deficits in dendritic L-VGCC calcium activity. Finding of DJ-1-regulated L-VGCC activity in dendrites in TSC and AD provides a unique signaling pathway that can be targeted in clinical mTORopathies.


Subject(s)
Alzheimer Disease , Tuberous Sclerosis , Animals , Mice , Alzheimer Disease/genetics , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Dendrites/metabolism , Mammals/metabolism , Tuberous Sclerosis/genetics
2.
Eur J Neurosci ; 57(8): 1241-1259, 2023 04.
Article in English | MEDLINE | ID: mdl-36840503

ABSTRACT

Alcohol use disorder (AUD) and anxiety/stressor disorders frequently co-occur and this dual diagnosis represents a major health and economic problem worldwide. The basolateral amygdala (BLA) is a key brain region that is known to contribute to the aetiology of both disorders. Although many studies have implicated BLA hyperexcitability in the pathogenesis of AUD and comorbid conditions, relatively little is known about the specific efferent projections from this brain region that contribute to these disorders. Recent optogenetic studies have shown that the BLA sends a strong monosynaptic excitatory projection to the ventral hippocampus (vHC) and that this circuit modulates anxiety- and fear-related behaviours. However, it is not known if this pathway influences alcohol drinking-related behaviours. Here, we employed a rodent operant self-administration regimen that procedurally separates appetitive (e.g. seeking) and consummatory (e.g., drinking) behaviours, chemogenetics and brain region-specific microinjections, to determine if BLA-vHC circuitry influences alcohol and sucrose drinking-related measures. We first confirmed prior optogenetic findings that silencing this circuit reduced anxiety-like behaviours on the elevated plus maze. We then demonstrated that inhibiting the BLA-vHC pathway significantly reduced appetitive drinking-related behaviours for both alcohol and sucrose while having no effect on consummatory measures. Taken together, these findings provide the first indication that the BLA-vHC circuit may regulate appetitive reward seeking directed at alcohol and natural rewards and add to a growing body of evidence suggesting that dysregulation of this pathway may contribute to the pathophysiology of AUD and anxiety/stressor-related disorders.


Subject(s)
Alcoholism , Basolateral Nuclear Complex , Humans , Hippocampus , Ethanol/pharmacology , Alcohol Drinking , Sucrose/pharmacology
3.
Nurs Womens Health ; 27(2): 79-89, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36773627

ABSTRACT

OBJECTIVE: To improve rates of exclusive breastfeeding during the postpartum hospital stay by implementing a new role of dedicated neonatal assessment nurse (NAN), whose primary function was neonatal care beginning immediately after birth. DESIGN: Quality improvement project with plan-do-study-act using evidence-based guidelines for implementing the NAN role. SETTING/LOCAL PROBLEM: Labor and delivery department of a tertiary care teaching hospital in the southeastern United States; breastfeeding exclusivity rates at this hospital were in the range of 50%. PARTICIPANTS: Registered nurses employed in the labor-delivery-recovery unit, mother-baby unit, and NICU. INTERVENTION/MEASUREMENTS: The NAN role was implemented to promote immediate skin-to-skin care (SSC) for stable newborns after vaginal and cesarean birth. Each NAN's competency was evaluated at the beginning and end of the education session through a pretest/posttest, and a skills validation was used to affirm their readiness for the new role. The outcome measure was breastfeeding exclusivity at the time of discharge from the hospital. SSC initiation and duration immediately after birth were the process measures. RESULTS: Twenty-five bedside registered nurses participated in this quality improvement project. There was a statistically significant difference between the pretest and posttest scores (p < .001), indicating a knowledge increase. All nurses met the skills validation criteria. The rate of SSC immediately after vaginal birth increased from 49% to 82% and after cesarean birth from 33% to 63%. Breastfeeding exclusivity rate at the time of discharge from the hospital increased from 50% to 86%. CONCLUSION: The NAN role provided transitional care at the bedside without the separation of mothers and newborns. This was an innovative role, without the need to hire new staff, that provided evidence-based care, resulting in improved SSC and exclusivity of breastfeeding before discharge.


Subject(s)
Breast Feeding , Mother-Child Relations , Neonatal Nursing , Nurse's Role , Female , Humans , Infant, Newborn , Breast Feeding/statistics & numerical data , Neonatal Nursing/organization & administration , Quality Improvement , Nursing Evaluation Research
4.
Front Psychiatry ; 14: 1296527, 2023.
Article in English | MEDLINE | ID: mdl-38268565

ABSTRACT

Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. In the mature brain, inhibitory GABAergic signaling is critical in maintaining neuronal homeostasis and vital human behaviors such as cognition, emotion, and motivation. While classically known to inhibit neuronal function under physiological conditions, previous research indicates a paradoxical switch from inhibitory to excitatory GABAergic signaling that is implicated in several neurological disorders. Various mechanisms have been proposed to contribute to the excitatory switch such as chloride ion dyshomeostasis, alterations in inhibitory receptor expression, and modifications in GABAergic synaptic plasticity. Of note, the hypothesized mechanisms underlying excitatory GABAergic signaling are highlighted in a number of neurodevelopmental, substance use, stress, and neurodegenerative disorders. Herein, we present an updated review discussing the presence of excitatory GABAergic signaling in various neurological disorders, and their potential contributions towards disease pathology.

5.
eNeuro ; 9(6)2022.
Article in English | MEDLINE | ID: mdl-36280288

ABSTRACT

Nucleus basalis magnocellularis (NBM) cholinergic projections to the basolateral amygdala (BLA) regulate the acquisition and consolidation of fear-like and anxiety-like behaviors. However, it is unclear whether the alterations in the NBM-BLA circuit promote negative affect during ethanol withdrawal (WD). Therefore, we performed ex vivo whole-cell patch-clamp electrophysiology in both the NBM and the BLA of male Sprague Dawley rats following 10 d of chronic intermittent ethanol (CIE) exposure and 24 h of WD. We found that CIE exposure and withdrawal enhanced the neuronal excitability of NBM putative "cholinergic" neurons. We subsequently used optogenetics to directly manipulate NBM terminal activity within the BLA and measure cholinergic modulation of glutamatergic afferents and BLA pyramidal neurons. Our findings indicate that CIE and withdrawal upregulate NBM cholinergic facilitation of glutamate release via activation of presynaptic nicotinic acetylcholine receptors (AChRs). Ethanol withdrawal-induced increases in NBM terminal activity also enhance BLA pyramidal neuron firing. Collectively, our results provide a novel characterization of the NBM-BLA circuit and suggest that CIE-dependent modifications to NBM afferents enhance BLA pyramidal neuron activity during ethanol withdrawal.


Subject(s)
Basolateral Nuclear Complex , Substance Withdrawal Syndrome , Animals , Rats , Male , Ethanol/pharmacology , Rats, Sprague-Dawley , Amygdala/physiology , Basal Nucleus of Meynert
6.
J Vis Exp ; (185)2022 07 22.
Article in English | MEDLINE | ID: mdl-35867038

ABSTRACT

Synapses are the site of communication between neurons. Neuronal circuit strength is related to synaptic density, and the breakdown of synapses is characteristic of disease states like major depressive disorder (MDD) and Alzheimer's disease. Traditional techniques to investigate synapse numbers include genetic expression of fluorescent markers (e.g., green fluorescent protein (GFP)), dyes that fill a neuron (e.g., carbocyanine dye, DiI), and immunofluorescent detection of spine markers (e.g., postsynaptic density 95 (PSD95)). A major caveat to these proxy techniques is that they only identify postsynaptic changes. Yet, a synapse is a connection between a presynaptic terminal and a postsynaptic spine. The gold standard for measuring synapse formation/elimination requires time-consuming electron microscopy or array tomography techniques. These techniques require specialized training and costly equipment. Further, only a limited number of neurons can be assessed and are used to represent changes to an entire brain region. DetectSyn is a rapid fluorescent technique that identifies changes to synapse formation or elimination due to a disease state or drug activity. DetectSyn utilizes a rapid proximity ligation assay to detect juxtaposed pre- and postsynaptic proteins and standard fluorescent microscopy, a technique readily available to most laboratories. Fluorescent detection of the resulting puncta allows for quick and unbiased analysis of experiments. DetectSyn provides more representative results than electron microscopy because larger areas can be analyzed than a limited number of fluorescent neurons. Moreover, DetectSyn works for in vitro cultured neurons and fixed tissue slices. Finally, a method is provided to analyze the data collected from this technique. Overall, DetectSyn offers a procedure for detecting relative changes in synapse density across treatments or disease states and is more accessible than traditional techniques.


Subject(s)
Depressive Disorder, Major , Cells, Cultured , Coloring Agents/metabolism , Depressive Disorder, Major/metabolism , Hippocampus , Humans , Neurons/metabolism , Presynaptic Terminals/metabolism , Synapses/physiology
7.
Cell Rep ; 39(1): 110633, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385720

ABSTRACT

Dysregulation of biological rhythms plays a role in a wide range of psychiatric disorders. We report mechanistic insights into the rhythms of rapid dopamine signals and cholinergic interneurons (CINs) working in concert in the rodent striatum. These rhythms mediate diurnal variation in conditioned responses to reward-associated cues. We report that the dopamine signal-to-noise ratio varies according to the time of day and that phasic signals are magnified during the middle of the dark cycle in rats. We show that CINs provide the mechanism for diurnal variation in rapid dopamine signals by serving as a gain of function to the dopamine signal-to-noise ratio that adjusts across time of day. We also show that conditioned responses to reward-associated cues exhibit diurnal rhythms, with cue-directed behaviors observed exclusively midway through the dark cycle. We conclude that the rapid dopamine signaling rhythm is mediated by a diurnal rhythm in CIN activity, which influences learning and motivated behaviors across the time of day.


Subject(s)
Circadian Rhythm , Dopamine , Animals , Cholinergic Agents , Conditioning, Classical , Humans , Nucleus Accumbens/physiology , Rats , Reward
8.
Front Neurosci ; 15: 669075, 2021.
Article in English | MEDLINE | ID: mdl-33994940

ABSTRACT

Alcohol use disorder (AUD) differentially impacts men and women and a growing body of evidence points to sex-dependent adaptations in a number of brain regions. In a prior study, we explored the effect of a chronic intermittent ethanol exposure (CIE) model of AUD on neuronal and molecular adaptations in the dorsal and ventral domains of the hippocampus (dHC and vHC, respectively) in male rats. We found the vHC to be particularly sensitive to CIE, showing an increase in neuronal excitability and synaptic proteins associated with augmented excitation. These findings were accompanied by a CIE-dependent increase in anxiety-like behaviors. To explore sex-dependent adaptations in the hippocampus, we conducted a similar study in female rats. CIE-treated female rats showed a relatively modest increase in anxiety-like behaviors along with a robust increase in depressive-like measures. Despite both sexes showing clear evidence of a negative affective state following CIE, the vHC of females showed a decrease, rather than an increase, in neuronal excitability. In line with the reduced sensitivity to neural adaptations in the dHC of male rats, we were unable to identify any functional changes in the dHC of females. The functional changes of the vHC in female rats could not be explained by altered expression levels of a number of proteins typically associated with changes in neuronal excitability. Taken together, these findings point to sex as a major factor in CIE-dependent hippocampal adaptations that should be explored further to better understand possible gender differences in the etiology and treatment of AUD.

9.
Neuropsychopharmacology ; 46(10): 1724-1733, 2021 09.
Article in English | MEDLINE | ID: mdl-34040157

ABSTRACT

Increasing evidence suggests that females are more vulnerable to the harmful effects of drugs of abuse, including opioids. Additionally, rates of heroin-related deaths substantially increased in females from 1999 to 2017 [1], underscoring the need to evaluate sex differences in heroin vulnerability. Moreover, the neurobiological substrates underlying sexually dimorphic responding to heroin are not fully defined. Thus, we evaluated male and female Long Evans rats on acquisition, dose-responsiveness, and seeking for heroin self-administration (SA) as well as using a long access model to assess escalation of intake at low and high doses of heroin, 0.025 and 0.1 mg/kg/inf, respectively. We paired this with ex vivo fast-scan cyclic voltammetry (FSCV) in the medial nucleus accumbens (NAc) shell and quantification of mu-opioid receptor (MOR) protein in the ventral tegmental area (VTA) and NAc. While males and females had similar heroin SA acquisition rates, females displayed increased responding and intake across doses, seeking for heroin, and escalation on long access. However, we found that males and females had similar expression levels of MORs in the VTA and NAc, regardless of heroin exposure. FSCV results revealed that heroin exposure did not change single-pulse elicited dopamine release, but caused an increase in dopamine transporter activity in both males and females compared to their naïve counterparts. Phasic-like stimulations elicited robust increases in dopamine release in heroin-exposed females compared to heroin-naïve females, with no differences seen in males. Together, our results suggest that differential adaptations of dopamine terminals may underlie the increased heroin SA behaviors seen in females.


Subject(s)
Dopamine , Heroin , Animals , Female , Male , Nucleus Accumbens , Rats , Rats, Long-Evans , Self Administration
10.
Mol Psychiatry ; 26(6): 2350-2362, 2021 06.
Article in English | MEDLINE | ID: mdl-33432187

ABSTRACT

Rapid antidepressants are novel treatments for major depressive disorder (MDD) and work by blocking N-methyl-D-aspartate receptors (NMDARs), which, in turn, activate the protein synthesis pathway regulated by mechanistic/mammalian target of rapamycin complex 1 (mTORC1). Our recent work demonstrates that the RNA-binding protein Fragile X Mental Retardation Protein (FMRP) is downregulated in dendrites upon treatment with a rapid antidepressant. Here, we show that the behavioral effects of the rapid antidepressant Ro-25-6981 require FMRP expression, and treatment promotes differential mRNA binding to FMRP in an mTORC1-dependent manner. Further, these mRNAs are identified to regulate transsynaptic signaling. Using a novel technique, we show that synapse formation underlying the behavioral effects of Ro-25-6981 requires GABABR-mediated mTORC1 activity in WT animals. Finally, we demonstrate that in an animal model that lacks FMRP expression and has clinical relevance for Fragile X Syndrome (FXS), GABABR activity is detrimental to the effects of Ro-25-6981. These effects are rescued with the combined therapy of blocking GABABRs and NMDARs, indicating that rapid antidepressants alone may not be an effective treatment for people with comorbid FXS and MDD.


Subject(s)
Depressive Disorder, Major , Fragile X Syndrome , Animals , Antidepressive Agents/pharmacology , Depressive Disorder, Major/drug therapy , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Humans , Synapses
11.
Alcohol ; 91: 41-51, 2021 03.
Article in English | MEDLINE | ID: mdl-33321179

ABSTRACT

Early life stress is known to impact vulnerability to psychopathological disorders in adulthood, including anxiety and alcohol use disorder (AUD), but the mechanisms underlying susceptibility to these outcomes are not fully understood. In the current study, we used adolescent social isolation (ASI) to determine whether Heterogeneous Stock (HS) rats, an outbred model used for genetic fine-mapping, could be used to study the genetics contributing to ASI-induced anxiety- and AUD-like behavior. We isolated (ASI) or group-housed (adolescent group-housed; AGH) 64 male HS rats at 4 weeks of age. After 5 weeks in these housing conditions, multiple anxiety and coping/despair-like behaviors were measured. All rats were then individually housed and assessed for voluntary ethanol self-administration. At euthanasia, synaptoneurosomes were isolated from a subset of brains to examine the expression of two proteins associated with alcohol drinking-related behaviors, GluA1 and SK2, in the dorsal (dHC) and ventral hippocampus (vHC). We found that ASI increased hyperactivity in the open field test relative to AGH, with no changes in other anxiety-like behaviors. Surprisingly, ASI rats demonstrated decreased immobility and increased climbing in the forced swim test relative to AGH. In contrast to prior studies by us and others, we found no difference in self-administration of 20% ethanol, with decreased ethanol self-administration in ASI relative to AGH rats at higher ethanol concentrations. Furthermore, while ASI in Long-Evans rats resulted in decreased SK2 expression in vHC synaptosomes, no differences were seen in vHC synaptosomes for SK2 or GluA1 in HS rats. These results demonstrate that HS rats are protected against many of the negative effects previously seen in response to ASI, namely anxiety-like behavior and increased ethanol self-administration. The current work suggests that a lack of change in SK2 and GluA1 expression levels in the vHC may play a role in conferring this protection.


Subject(s)
Alcohol Drinking , Anxiety , Hyperkinesis/psychology , Stress, Psychological/complications , Animals , Ethanol , Male , Rats , Rats, Long-Evans
12.
J Nurs Care Qual ; 36(1): 50-56, 2021.
Article in English | MEDLINE | ID: mdl-32618811

ABSTRACT

BACKGROUND: Evidence suggests that a lack of sleep among patients during hospitalization can impact their health and well-being. LOCAL PROBLEM: On inpatient units, patients experience lack of sleep due to noise and multiple interruptions at night. METHODS: A pilot intervention to support the initiative, "Don't wake the patient for routine care unless the patient's condition or care specifically requires it," was implemented on 3 units in 3 hospitals. All the 3 units had experienced patient concerns about lack of sleep at night. INTERVENTIONS: Nurses implemented the project using a purposeful strategy of sleep masks, earplugs, noise detectors, and bundling care. RESULTS: Positive experiences of those patients who participated in the sleep intervention. CONCLUSIONS: There was multidisciplinary support to promote a better patient experience of nighttime sleep. The experiences across the 3 hospital units were positive for providers, patients, and patients' families.


Subject(s)
Sleep Wake Disorders , Sleep , Ear Protective Devices , Hospitalization , Humans , Noise
13.
ACS Chem Neurosci ; 10(4): 2033-2044, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30284806

ABSTRACT

Chronic peri-adolescent stress in humans increases risk to develop a substance use disorder during adulthood. Rats reared in social isolation during peri-adolescence (aSI; 1 rat/cage) period show greater ethanol and cocaine intake compared to group housed (aGH; 4 rats/cage) rats. In addition, aSI rats have a heightened dopamine response in the nucleus accumbens (NAc) to rewarding and aversive stimuli. Furthermore, single pulse electrical stimulation in slices containing NAc core elicits greater dopamine release in aSI rats. Here, we further investigated dopamine release kinetics and machinery following aSI. Dopamine release, across a wide range of stimulation intensities and frequencies, was significantly greater in aSI rats. Interestingly, subthreshold intensity stimulations also resulted in measurable dopamine release in accumbal slices from aSI but not aGH rats. Extracellular [Ca2+] manipulations revealed augmented calcium sensitivity of dopamine release in aSI rats. The readily releasable pools of dopamine, examined by bath application of Ro-04-1284/000, a vesicular monoamine transporter 2 (VMAT2) inhibitor, were depleted faster in aGH rats. Western blot analysis of release machinery proteins (VMAT2, Synaptogyrin-3, Syntaxin-1, and Munc13-3) showed no difference between the two groups. Tyrosine hydroxylase (TH) protein expression levels, however, were elevated in aSI rats. The greater dopamine release could potentially be explained by higher levels of TH, the rate-limiting step for dopamine synthesis. This augmented responsivity of the dopamine system and heightened dopamine availability post-aSI may lead to an increased risk of addiction vulnerability.


Subject(s)
Dopamine/metabolism , Nucleus Accumbens/metabolism , Presynaptic Terminals/metabolism , Social Isolation , Stress, Psychological/metabolism , Vesicular Monoamine Transport Proteins/metabolism , Age Factors , Animals , Chronic Disease , Dopamine Uptake Inhibitors/pharmacology , Male , Nucleus Accumbens/drug effects , Presynaptic Terminals/drug effects , Rats , Rats, Long-Evans , Social Isolation/psychology , Stress, Psychological/psychology , Vesicular Monoamine Transport Proteins/antagonists & inhibitors
14.
Neuroscience ; 398: 144-157, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30481568

ABSTRACT

Many studies have implicated hippocampal dysregulation in the pathophysiology of alcohol use disorder (AUD). However, over the past twenty years, a growing body of evidence has revealed distinct functional roles of the dorsal (dHC) and ventral (vHC) hippocampal subregions, with the dHC being primarily involved in spatial learning and memory and the vHC regulating anxiety- and depressive-like behaviors. Notably, to our knowledge, no rodent studies have examined the effects of chronic ethanol exposure on synaptic transmission along the dorsal/ventral axis. To that end, we examined the effects of the chronic intermittent ethanol vapor exposure (CIE) model of AUD on dHC and vHC synaptic excitability. Adult male Long-Evans rats were exposed to CIE or AIR for 10 days (12 h/day; targeting blood ethanol levels of 175-225 mg%) and recordings were made 24 h into withdrawal. As expected, this protocol increased anxiety-like behaviors on the elevated plus-maze and successive alleys test. Extracellular recordings revealed marked CIE-associated increases in synaptic excitation in the CA1 region that were exclusively restricted to the ventral domain of the hippocampus. Western blot analysis of synaptoneurosomal fractions revealed that the expression of two proteins that regulate synaptic strength, GluA2 and SK2, were dysregulated in the vHC, but not the dHC, following CIE. Together, these findings suggest that the ventral CA1 region may be particularly sensitive to the maladaptive effects of chronic ethanol exposure and provide new insight into some of the neural substrates that may contribute to the negative affective state that develops during withdrawal.


Subject(s)
Alcohol-Related Disorders/physiopathology , Central Nervous System Depressants/adverse effects , Ethanol/adverse effects , Hippocampus/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Animals , Disease Models, Animal , Gene Expression/drug effects , Germinal Center Kinases , Hippocampus/physiopathology , Male , Protein Serine-Threonine Kinases/metabolism , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Synaptic Transmission/physiology
15.
Neuropharmacology ; 146: 289-299, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30419244

ABSTRACT

Alcohol use disorder (AUD) and major depressive disorder (MDD) are prevalent, debilitating, and highly comorbid disorders. The molecular changes that underlie their comorbidity are beginning to emerge. For example, recent evidence showed that acute ethanol exposure produces rapid antidepressant-like biochemical and behavioral responses. Both ethanol and fast-acting antidepressants block N-methyl-D-aspartate receptor (NMDAR) activity, leading to synaptic changes and long-lasting antidepressant-like behavioral effects. We used RNA sequencing to analyze changes in the synaptic transcriptome after acute treatment with ethanol or the NMDAR antagonist, Ro 25-6981. Ethanol and Ro 25-6981 induced differential, independent changes in gene expression. In contrast with gene-level expression, ethanol and Ro 25-6981 produced overlapping changes in exons, as measured by analysis of differentially expressed exons (DEEs). A prominent overlap in genes with DEEs indicated that changes in exon usage were important for both ethanol and Ro 25-6981 action. Structural modeling provided evidence that ethanol-induced exon expression in the NMDAR1 amino-terminal domain could induce conformational changes and thus alter NMDAR function. These findings suggest that the rapid antidepressant effects of ethanol and NMDAR antagonists reported previously may depend on synaptic exon usage rather than gene expression.


Subject(s)
Alcoholism/genetics , Depressive Disorder, Major/genetics , Exons/drug effects , Exons/genetics , Gene Expression/drug effects , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Antidepressive Agents/pharmacology , Comorbidity , Ethanol/pharmacology , Hippocampus/drug effects , Male , Mice, Inbred C57BL , Models, Animal , Phenols/pharmacology , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Neurotransmitter , Transcriptome
16.
J Nurs Care Qual ; 33(2): 157-165, 2018.
Article in English | MEDLINE | ID: mdl-28658191

ABSTRACT

This study explored the trajectory of patients who remained on a general unit after medical emergency team activation. Of those who had a second activation within 24 hours, 80% occurred within 12 hours of the baseline activation. Chest pain and recent intensive care unit discharge were associated with having a second activation. There were statistically, not clinically, significant associations between mean vital signs and second activations; however, the patterns of change may be clinically useful.


Subject(s)
Hospital Rapid Response Team , Intensive Care Units , Patient Admission , Chest Pain/etiology , Electronic Health Records/statistics & numerical data , Female , Humans , Longitudinal Studies , Male , Patient Discharge , Prospective Studies , Time Factors , Vital Signs/physiology
17.
Front Cell Neurosci ; 11: 152, 2017.
Article in English | MEDLINE | ID: mdl-28611595

ABSTRACT

The mammalian/mechanistic target of rapamycin complex 1 (mTORC1) serves as a regulator of mRNA translation. Recent studies suggest that mTORC1 may also serve as a local, voltage sensor in the postsynaptic region of neurons. Considering biochemical, bioinformatics and imaging data, we hypothesize that the activity state of mTORC1 dynamically regulates local membrane potential by promoting and repressing protein synthesis of select mRNAs. Our hypothesis suggests that mTORC1 uses positive and negative feedback pathways, in a branch-specific manner, to maintain neuronal excitability within an optimal range. In some dendritic branches, mTORC1 activity oscillates between the "On" and "Off" states. We define this as negative feedback. In contrast, positive feedback is defined as the pathway that leads to a prolonged depolarized or hyperpolarized resting membrane potential, whereby mTORC1 activity is constitutively on or off, respectively. We propose that inactivation of mTORC1 increases the expression of voltage-gated potassium alpha (Kv1.1 and 1.2) and beta (Kvß2) subunits, ensuring that the membrane resets to its resting membrane potential after experiencing increased synaptic activity. In turn, reduced mTORC1 activity increases the protein expression of syntaxin-1A and promotes the surface expression of the ionotropic glutamate receptor N-methyl-D-aspartate (NMDA)-type subunit 1 (GluN1) that facilitates increased calcium entry to turn mTORC1 back on. Under conditions such as learning and memory, mTORC1 activity is required to be high for longer periods of time. Thus, the arm of the pathway that promotes syntaxin-1A and Kv1 protein synthesis will be repressed. Moreover, dendritic branches that have low mTORC1 activity with increased Kv expression would balance dendrites with constitutively high mTORC1 activity, allowing for the neuron to maintain its overall activity level within an ideal operating range. Finally, such a model suggests that recruitment of more positive feedback dendritic branches within a neuron is likely to lead to neurodegenerative disorders.

18.
FEBS Lett ; 591(11): 1540-1554, 2017 06.
Article in English | MEDLINE | ID: mdl-28493559

ABSTRACT

Mammalian target of rapamycin (mTOR) activity is required for memory and is dysregulated in disease. Activation of mTOR promotes protein synthesis; however, new studies are demonstrating that mTOR activity also represses the translation of mRNAs. Almost three decades ago, Kandel and colleagues hypothesised that memory was due to the induction of positive regulators and removal of negative constraints. Are these negative constraints repressed mRNAs that code for proteins that block memory formation? Herein, we will discuss the mRNAs coded by putative memory suppressors, how activation/inactivation of mTOR repress protein expression at the synapse, how mTOR activity regulates RNA binding proteins, mRNA stability, and translation, and what the possible implications of mRNA repression are to memory and neurodegenerative disorders.


Subject(s)
Memory/physiology , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Humans , Neurodegenerative Diseases/metabolism , RNA-Binding Proteins/metabolism , Synapses/metabolism
19.
Front Mol Neurosci ; 10: 45, 2017.
Article in English | MEDLINE | ID: mdl-28286470

ABSTRACT

In the last decade, bioinformatic analyses of high-throughput proteomics and transcriptomics data have enabled researchers to gain insight into the molecular networks that may underlie lasting changes in synaptic efficacy. Development and utilization of these techniques have advanced the field of learning and memory significantly. It is now possible to move from the study of activity-dependent changes of a single protein to modeling entire network changes that require local protein synthesis. This data revolution has necessitated the development of alternative computational and statistical techniques to analyze and understand the patterns contained within. Thus, the focus of this review is to provide a synopsis of the journey and evolution toward big data techniques to address still unanswered questions regarding how synapses are modified to strengthen neuronal circuits. We first review the seminal studies that demonstrated the pivotal role played by local mRNA translation as the mechanism underlying the enhancement of enduring synaptic activity. In the interest of those who are new to the field, we provide a brief overview of molecular biology and biochemical techniques utilized for sample preparation to identify locally translated proteins using RNA sequencing and proteomics, as well as the computational approaches used to analyze these data. While many mRNAs have been identified, few have been shown to be locally synthesized. To this end, we review techniques currently being utilized to visualize new protein synthesis, a task that has proven to be the most difficult aspect of the field. Finally, we provide examples of future applications to test the physiological relevance of locally synthesized proteins identified by big data approaches.

20.
Nat Commun ; 7: 12867, 2016 Sep 26.
Article in English | MEDLINE | ID: mdl-27666021

ABSTRACT

Alcohol promotes lasting neuroadaptive changes that may provide relief from depressive symptoms, often referred to as the self-medication hypothesis. However, the molecular/synaptic pathways that are shared by alcohol and antidepressants are unknown. In the current study, acute exposure to ethanol produced lasting antidepressant and anxiolytic behaviours. To understand the functional basis of these behaviours, we examined a molecular pathway that is activated by rapid antidepressants. Ethanol, like rapid antidepressants, alters γ-aminobutyric acid type B receptor (GABABR) expression and signalling, to increase dendritic calcium. Furthermore, new GABABRs are synthesized in response to ethanol treatment, requiring fragile-X mental retardation protein (FMRP). Ethanol-dependent changes in GABABR expression, dendritic signalling, and antidepressant efficacy are absent in Fmr1-knockout (KO) mice. These findings indicate that FMRP is an important regulator of protein synthesis following alcohol exposure, providing a molecular basis for the antidepressant efficacy of acute ethanol exposure.

SELECTION OF CITATIONS
SEARCH DETAIL
...