Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 883
1.
Transl Psychiatry ; 14(1): 200, 2024 May 07.
Article En | MEDLINE | ID: mdl-38714646

Lithium is an effective augmenting agent for depressed patients with inadequate response to standard antidepressant therapy, but numerous adverse effects limit its use. We previously reported that a lithium-mimetic agent, ebselen, promoted a positive emotional bias-an indicator of potential antidepressant activity in healthy participants. We therefore aimed to investigate the effects of short-term ebselen treatment on emotional processing and brain neurochemistry in depressed patients with inadequate response to standard antidepressants. We conducted a double-blind, placebo-controlled 7-day experimental medicine study in 51 patients with major depressive disorder who were currently taking antidepressants but had an inadequate response to treatment. Participants received either ebselen 600 mg twice daily for seven days or identical matching placebo. An emotional testing battery, magnetic resonance spectroscopy and depression and anxiety rating scales were conducted at baseline and after seven days of treatment. Ebselen did not increase the recognition of positive facial expressions in the depressed patient group. However, ebselen increased the response bias towards fear emotion in the signal detection measurement. In the anterior cingulate cortex, ebselen significantly reduced the concentrations of inositol and Glx (glutamate+glutamine). We found no significant differences in depression and anxiety rating scales between visits. Our study did not find any positive shift in emotional bias in depressed patients with an inadequate response to antidepressant medication. We confirmed the ability of ebselen to lower inositol and Glx in the anterior cingulate cortex. These latter effects are probably mediated through inhibition of inositol monophosphatase and glutaminase respectively.


Antidepressive Agents , Azoles , Depressive Disorder, Major , Emotions , Isoindoles , Organoselenium Compounds , Humans , Female , Male , Organoselenium Compounds/pharmacology , Double-Blind Method , Adult , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Antidepressive Agents/therapeutic use , Antidepressive Agents/pharmacology , Middle Aged , Emotions/drug effects , Azoles/pharmacology , Magnetic Resonance Spectroscopy , Depressive Disorder, Treatment-Resistant/drug therapy , Depressive Disorder, Treatment-Resistant/metabolism , Gyrus Cinguli/metabolism , Gyrus Cinguli/drug effects , Gyrus Cinguli/diagnostic imaging , Brain/drug effects , Brain/metabolism , Brain/diagnostic imaging
2.
Mol Pain ; 20: 17448069241258110, 2024.
Article En | MEDLINE | ID: mdl-38744422

Recent studies using different experimental approaches demonstrate that silent synapses may exist in the adult cortex including the sensory cortex and anterior cingulate cortex (ACC). The postsynaptic form of long-term potentiation (LTP) in the ACC recruits some of these silent synapses and the activity of calcium-stimulated adenylyl cyclases (ACs) is required for such recruitment. It is unknown if the chemical activation of ACs may recruit silent synapses. In this study, we found that activation of ACs contributed to synaptic potentiation in the ACC of adult mice. Forskolin, a selective activator of ACs, recruited silent responses in the ACC of adult mice. The recruitment was long-lasting. Interestingly, the effect of forskolin was not universal, some silent synapses did not undergo potentiation or recruitment. These findings suggest that these adult cortical synapses are not homogenous. The application of a selective calcium-permeable AMPA receptor inhibitor 1-naphthyl acetyl spermine (NASPM) reversed the potentiation and the recruitment of silent responses, indicating that the AMPA receptor is required. Our results strongly suggest that the AC-dependent postsynaptic AMPA receptor contributes to the recruitment of silent responses at cortical LTP.


Adenylyl Cyclases , Colforsin , Gyrus Cinguli , Long-Term Potentiation , Animals , Mice , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Colforsin/pharmacology , Long-Term Potentiation/drug effects , Adenylyl Cyclases/metabolism , Male , Receptors, AMPA/metabolism , Mice, Inbred C57BL , Synapses/drug effects , Synapses/metabolism , Calcium/metabolism
3.
Transl Psychiatry ; 14(1): 211, 2024 May 27.
Article En | MEDLINE | ID: mdl-38802372

Lamotrigine is an effective mood stabiliser, largely used for the management and prevention of depression in bipolar disorder. The neuropsychological mechanisms by which lamotrigine acts to relieve symptoms as well as its neural effects on emotional processing remain unclear. The primary objective of this current study was to investigate the impact of an acute dose of lamotrigine on the neural response to a well-characterised fMRI task probing implicit emotional processing relevant to negative bias. 31 healthy participants were administered either a single dose of lamotrigine (300 mg, n = 14) or placebo (n = 17) in a randomized, double-blind design. Inside the 3 T MRI scanner, participants completed a covert emotional faces gender discrimination task. Brain activations showing significant group differences were identified using voxel-wise general linear model (GLM) nonparametric permutation testing, with threshold free cluster enhancement (TFCE) and a family wise error (FWE)-corrected cluster significance threshold of p < 0.05. Participants receiving lamotrigine were more accurate at identifying the gender of fearful (but not happy or angry) faces. A network of regions associated with emotional processing, including amygdala, insula, and the anterior cingulate cortex (ACC), was significantly less activated in the lamotrigine group compared to the placebo group across emotional facial expressions. A single dose of lamotrigine reduced activation in limbic areas in response to faces with both positive and negative expressions, suggesting a valence-independent effect. However, at a behavioural level lamotrigine appeared to reduce the distracting effect of fear on face discrimination. Such effects may be relevant to the mood stabilisation effects of lamotrigine.


Emotions , Facial Expression , Healthy Volunteers , Lamotrigine , Magnetic Resonance Imaging , Triazines , Humans , Lamotrigine/pharmacology , Lamotrigine/administration & dosage , Male , Female , Adult , Double-Blind Method , Emotions/drug effects , Triazines/pharmacology , Triazines/administration & dosage , Young Adult , Brain/drug effects , Brain/diagnostic imaging , Facial Recognition/drug effects , Gyrus Cinguli/drug effects , Gyrus Cinguli/diagnostic imaging , Amygdala/drug effects , Amygdala/diagnostic imaging , Antimanic Agents/pharmacology , Antimanic Agents/administration & dosage
4.
Neuroimage ; 293: 120619, 2024 Jun.
Article En | MEDLINE | ID: mdl-38679186

Catecholamines and amino acid transmitter systems are known to interact, the exact links and their impact on cognitive control functions have however remained unclear. Using a multi-modal imaging approach combining EEG and proton-magnetic resonance spectroscopy (1H-MRS), we investigated the effect of different degrees of pharmacological catecholaminergic enhancement onto theta band activity (TBA) as a measure of interference control during response inhibition and execution. It was central to our study to evaluate the predictive impact of in-vivo baseline GABA+ concentrations in the striatum, the anterior cingulate cortex (ACC) and the supplemental motor area (SMA) of healthy adults under varying degrees of methylphenidate (MPH) stimulation. We provide evidence for a predictive interrelation of baseline GABA+ concentrations in cognitive control relevant brain areas onto task-induced TBA during response control stimulated with MPH. Baseline GABA+ concentrations in the ACC, the striatum, and the SMA had a differential impact on predicting interference control-related TBA in response execution trials. GABA+ concentrations in the ACC appeared to be specifically important for TBA modulations when the cognitive effort needed for interference control was high - that is when no prior task experience exists, or in the absence of catecholaminergic enhancement with MPH. The study highlights the predictive role of baseline GABA+ concentrations in key brain areas influencing cognitive control and responsiveness to catecholaminergic enhancement, particularly in high-effort scenarios.


Catecholamines , Cognition , Electroencephalography , Methylphenidate , Proton Magnetic Resonance Spectroscopy , gamma-Aminobutyric Acid , Humans , gamma-Aminobutyric Acid/metabolism , Male , Adult , Female , Young Adult , Proton Magnetic Resonance Spectroscopy/methods , Catecholamines/metabolism , Methylphenidate/pharmacology , Electroencephalography/methods , Cognition/physiology , Brain/metabolism , Brain/diagnostic imaging , Gyrus Cinguli/metabolism , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/drug effects , Theta Rhythm/physiology , Theta Rhythm/drug effects , Executive Function/physiology , Executive Function/drug effects , Central Nervous System Stimulants/pharmacology
5.
Neuroimage ; 249: 118887, 2022 04 01.
Article En | MEDLINE | ID: mdl-34999203

An essential core function of one's cognitive flexibility is the use of acquired knowledge and skills to adapt to ongoing environmental changes. Animal models have highlighted the influence serotonin has on neuroplasticity. These effects have been predominantly demonstrated during emotional relearning which is theorized as a possible model for depression. However, translation of these mechanisms is in its infancy. To this end, we assessed changes in effective connectivity at rest and during associative learning as a proxy of neuroplastic changes in healthy volunteers. 76 participants underwent 6 weeks of emotional or non-emotional (re)learning (face-matching or Chinese character-German noun matching). During relearning participants either self-administered 10 mg/day of the selective serotonin reuptake inhibitor (SSRI) escitalopram or placebo in a double-blind design. Associative learning tasks, resting-state and structural images were recorded before and after both learning phases (day 1, 21 and 42). Escitalopram intake modulated relearning changes in a network encompassing the right insula, anterior cingulate cortex and right angular gyrus. Here, the process of relearning during SSRI intake showed a greater decrease in effective connectivity from the right insula to both the anterior cingulate cortex and right angular gyrus, with increases in the opposite direction when compared to placebo. In contrast, intrinsic connections and those at resting-state were only marginally affected by escitalopram. Further investigation of gray matter volume changes in these functionally active regions revealed no significant SSRI-induced structural changes. These findings indicate that the right insula plays a central role in the process of relearning and SSRIs further potentiate this effect. In sum, we demonstrated that SSRIs amplify learning-induced effective connections rather than affecting the intrinsic task connectivity or that of resting-state.


Association Learning , Connectome , Insular Cortex , Nerve Net , Neuronal Plasticity , Selective Serotonin Reuptake Inhibitors/pharmacology , Adult , Association Learning/drug effects , Association Learning/physiology , Citalopram/pharmacology , Female , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/drug effects , Gyrus Cinguli/physiology , Humans , Insular Cortex/diagnostic imaging , Insular Cortex/drug effects , Insular Cortex/physiology , Magnetic Resonance Imaging , Male , Nerve Net/diagnostic imaging , Nerve Net/drug effects , Nerve Net/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Parietal Lobe/diagnostic imaging , Parietal Lobe/drug effects , Parietal Lobe/physiology , Rest , Selective Serotonin Reuptake Inhibitors/administration & dosage , Young Adult
6.
Addict Biol ; 27(1): e13085, 2022 01.
Article En | MEDLINE | ID: mdl-34390300

Disrupted brain gamma-aminobutyric acid (GABA)/glutamate homeostasis is a promising target for pharmacological intervention in co-occurring bipolar disorder (BD) and cannabis use disorder (CUD). Gabapentin is a safe and well-tolerated medication, FDA-approved to treat other neurological diseases, that restores GABA/glutamate homeostasis, with treatment studies supporting efficacy in treating CUD, as well as anxiety and sleep disorders that are common to both BD and CUD. The present manuscript represents the primary report of a randomized, double-blind, placebo-controlled, crossover (1-week/condition), multimodal-MRI (proton-MR spectroscopy, functional MRI) pilot study of gabapentin (1200 mg/day) in BD + CUD (n = 22). Primary analyses revealed that (1) gabapentin was well tolerated and adherence and retention were high, (2) gabapentin increased dorsal anterior cingulate cortex (dACC) and right basal ganglia (rBG) glutamate levels and (3) gabapentin increased activation to visual cannabis cues in the posterior midcingulate cortex (pMCC, a region involved in response inhibition to rewarding stimuli). Exploratory evaluation of clinical outcomes further found that in participants taking gabapentin versus placebo, (1) elevations of dACC GABA levels were associated with lower manic/mixed and depressive symptoms and (2) elevations of rBG glutamate levels and pMCC activation to cannabis cues were associated with lower cannabis use. Though promising, the findings from this study should be interpreted with caution due to observed randomization order effects on dACC glutamate levels and identification of statistical moderators that differed by randomization order (i.e. cigarette-smoking status on rBG glutamate levels and pMCC cue activation). Nonetheless, they provide the necessary foundation for a more robustly designed (urn-randomized, parallel-group) future study of adjuvant gabapentin for BD + CUD.


Bipolar Disorder/drug therapy , Gabapentin/therapeutic use , Glutamic Acid/drug effects , Marijuana Abuse/drug therapy , gamma-Aminobutyric Acid/drug effects , Adolescent , Adult , Bipolar Disorder/epidemiology , Cigarette Smoking/epidemiology , Double-Blind Method , Female , Gabapentin/administration & dosage , Gabapentin/adverse effects , Gyrus Cinguli/drug effects , Humans , Magnetic Resonance Imaging , Male , Marijuana Abuse/epidemiology , Middle Aged , Pilot Projects , Proton Magnetic Resonance Spectroscopy , Young Adult
7.
Article En | MEDLINE | ID: mdl-34971723

BACKGROUND: Sub-anesthetic ketamine doses rapidly reduce depressive symptoms, although additional investigations of the underlying neural mechanisms and the prediction of response outcomes are needed. Electroencephalographic (EEG)-derived measures have shown promise in predicting antidepressant response to a variety of treatments, and are sensitive to ketamine administration. This study examined their utility in characterizing changes in depressive symptoms following single and repeated ketamine infusions. METHODS: Recordings were obtained from patients with treatment-resistant major depressive disorder (MDD) (N = 24) enrolled in a multi-phase clinical ketamine trial. During the randomized, double-blind, crossover phase (Phase 1), patients received intravenous ketamine (0.5 mg/kg) and midazolam (30 µg/kg), at least 1 week apart. For each medication, three resting, eyes-closed recordings were obtained per session (pre-infusion, immediately post-infusion, 2 h post-infusion), and changes in power (delta, theta1/2/total, alpha1/2/total, beta, gamma), alpha asymmetry, theta cordance, and theta source-localized anterior cingulate cortex activity were quantified. The relationships between ketamine-induced changes with early (Phase 1) and sustained (Phases 2,3: open-label repeated infusions) decreases in depressive symptoms (Montgomery-Åsberg Depression Rating Score, MADRS) and suicidal ideation (MADRS item 10) were examined. RESULTS: Both medications decreased alpha and theta immediately post-infusion, however, only midazolam increased delta (post-infusion), and only ketamine increased gamma (immediately post- and 2 h post-infusion). Regional- and frequency-specific ketamine-induced EEG changes were related to and predictive of decreases in depressive symptoms (theta, gamma) and suicidal ideation (alpha). Early and sustained treatment responders differed at baseline in surface-level and source-localized theta. CONCLUSIONS: Ketamine exerts frequency-specific changes on EEG-derived measures, which are related to depressive symptom decreases in treatment-resistant MDD and provide information regarding early and sustained individual response to ketamine. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov: Action of Ketamine in Treatment-Resistant Depression, NCT01945047.


Analgesics/administration & dosage , Brain Waves/drug effects , Depressive Disorder, Treatment-Resistant/drug therapy , Electrophysiology , Gyrus Cinguli/drug effects , Ketamine/administration & dosage , Adult , Anesthetics, Intravenous/administration & dosage , Cross-Over Studies , Electroencephalography , Female , Humans , Infusions, Intravenous , Male , Midazolam/administration & dosage , Psychiatric Status Rating Scales
8.
Sci Rep ; 11(1): 24267, 2021 12 20.
Article En | MEDLINE | ID: mdl-34930953

The neurobiological basis of brand and product attachment has received much attention in consumer neuroscience research, although it remains unclear. In this study, we conducted functional MRI experiments involving female users of famous luxury brand cosmetics as participants, based on the regions of interest involved in human attachment and object attachment. The results showed that the left ventral pallidum (VP), which is involved in positive reward, and the right posterior cingulate cortex (PCC), which is involved in self-concept, a key concept in object attachment, are the core regions in cosmetic attachment. Moreover, the performed psychophysiological interaction analyses showed that VP-temporoparietal junction connectivity positively correlated with activity in the dorsal raphe nucleus, and PCC-anterior hippocampus (aHC) connectivity positively correlated with subjective evaluation of attachment. The former suggests that object attachment is a human-like attachment and a stronger tendency of anthropomorphism is associated with stronger feelings of security. The latter suggests that the individual's concept of attachment as well as the relationships with the attached cosmetics are represented in the aHC, and the PCC-aHC associations produce subjective awareness of the attachment relationships. These associations between memory and reward systems have been shown to play critical roles in cosmetic attachment.


Attention/physiology , Basal Forebrain/drug effects , Cosmetics/chemistry , Gyrus Cinguli/drug effects , Neurons/metabolism , Adult , Basal Forebrain/diagnostic imaging , Brain/diagnostic imaging , Brain Mapping , Female , Gyrus Cinguli/diagnostic imaging , Humans , Magnetic Resonance Imaging , Models, Neurological , Nerve Net/physiology , Neural Pathways/physiopathology , Object Attachment , Regression Analysis , Touch
9.
Cell Rep ; 37(9): 109933, 2021 11 30.
Article En | MEDLINE | ID: mdl-34852233

Pyramidal neurons in the anterior cingulate cortex (ACC), a prefrontal region involved in processing the affective components of pain, display hyperexcitability in chronic neuropathic pain conditions, and their silencing abolishes hyperalgesia. We show that dopamine, through D1 receptor (D1R) signaling, inhibits pyramidal neurons of mouse ACC by modulation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Activation of Gs-coupled D1R by dopamine induces the opening of HCN channels at physiological membrane potentials, driving a significant decrease in input resistance and excitability. Systemic L-DOPA in chronic neuropathic mice rescues HCN channel activity, normalizes pyramidal excitability in ACC, and blocks mechanical and thermal allodynia. Moreover, microinjection of a selective D1R agonist in the ACC relieves the aversiveness of ongoing neuropathic pain, while an ACC D1R antagonist blocks gabapentin- and lidocaine-evoked antinociception. We conclude that dopaminergic inhibition via D1R in ACC plays an analgesic role in physiological conditions and is decreased in chronic pain.


Dopamine/metabolism , Gyrus Cinguli/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Levodopa/pharmacology , Neuralgia/prevention & control , Potassium Channels/metabolism , Pyramidal Cells/drug effects , Receptors, Dopamine D1/agonists , Animals , Dopamine Agents/pharmacology , Gyrus Cinguli/metabolism , Gyrus Cinguli/pathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Male , Membrane Potentials , Neuralgia/etiology , Neuralgia/metabolism , Neuralgia/pathology , Potassium Channels/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Rats, Sprague-Dawley
10.
Mol Brain ; 14(1): 140, 2021 09 15.
Article En | MEDLINE | ID: mdl-34526080

Previous studies have demonstrated that brain-derived neurotrophic factor (BDNF) is one of the diffusible messengers for enhancing synaptic transmission in the hippocampus. Less information is available about the possible roles of BDNF in the anterior cingulate cortex (ACC). In the present study, we used 64-electrode array field recording system to investigate the effect of BDNF on ACC excitatory transmission. We found that BDNF enhanced synaptic responses in a dose-dependent manner in the ACC in C57/BL6 mice. The enhancement was long-lasting, and persisted for at least 3 h. In addition to the enhancement, BDNF also recruited inactive synaptic responses in the ACC. Bath application of the tropomyosin receptor kinase B (TrkB) receptor antagonist K252a blocked BDNF-induced enhancement. L-type voltage-gated calcium channels (L-VGCC), metabotropic glutamate receptors (mGluRs), but not NMDA receptors were required for BDNF-produced enhancement. Moreover, calcium-stimulated adenylyl cyclase subtype 1 (AC1) but not AC8 was essential for the enhancement. A selective AC1 inhibitor NB001 completely blocked the enhancement. Furthermore, BDNF-produced enhancement occluded theta burst stimulation (TBS) induced long-term potentiation (LTP), suggesting that they may share similar signaling mechanisms. Finally, the expression of BDNF-induced enhancement depends on postsynaptic incorporation of calcium-permeable AMPA receptors (CP-AMPARs) and protein kinase Mζ (PKMζ). Our results demonstrate that cortical BDNF may contribute to synaptic potentiation in the ACC.


Brain-Derived Neurotrophic Factor/pharmacology , Gyrus Cinguli/drug effects , Long-Term Potentiation/drug effects , Synapses/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adenylyl Cyclases/physiology , Animals , Calcium Channels, L-Type/physiology , Carbazoles/pharmacology , Dose-Response Relationship, Drug , Electrodes, Implanted , Indole Alkaloids/pharmacology , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Protein Kinase C/physiology , Receptors, AMPA/physiology , Receptors, Metabotropic Glutamate/physiology , Synapses/physiology , Theta Rhythm/drug effects
11.
Neurosci Lett ; 764: 136205, 2021 11 01.
Article En | MEDLINE | ID: mdl-34478818

Lactate transport is an important means of communication between astrocytes and neurons and is implicated in a variety of neurobiological processes. However, the connection between astrocyte-neuron lactate transport and nociceptive modulation has not been well established. Here, we found that Complete Freund's adjuvant (CFA)-induced inflammation pain leads to a significant increase in extracellular lactate levels in the anterior cingulate cortex (ACC). Inhibition of glycogenolysis and lactate release in the ACC disrupted the persistent, but not acute, inflammation pain induced by CFA, and this effect was reversed by exogenous L-lactate administration. Knocking down the expression of lactate transporters (MCT1, MCT4, or MCT2) also disrupted the long lasting inflammation pain induced by CFA. Moreover, glycogenolysis in the ACC is critical for the induction of molecular changes related to neuronal plasticity, including the induction of phospho- (p-) ERK, p-CREB, and Fos. Taken together, our findings indicate that astrocyte-neuron lactate transport in the ACC is critical for the occurrence of persistent inflammation pain, suggesting a novel mechanism underlying chronic pain.


Arabinose/pharmacology , Cell Communication/immunology , Chronic Pain/immunology , Gyrus Cinguli/pathology , Imino Furanoses/pharmacology , Lactic Acid/metabolism , Sugar Alcohols/pharmacology , Animals , Arabinose/therapeutic use , Astrocytes/metabolism , Cell Communication/drug effects , Chronic Pain/drug therapy , Chronic Pain/pathology , Disease Models, Animal , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Glycogenolysis/drug effects , Glycogenolysis/immunology , Gyrus Cinguli/cytology , Gyrus Cinguli/drug effects , Gyrus Cinguli/immunology , Humans , Imino Furanoses/therapeutic use , Male , Mice , Neuronal Plasticity/drug effects , Neuronal Plasticity/immunology , Neurons/metabolism , Sugar Alcohols/therapeutic use
12.
Neuropharmacology ; 197: 108749, 2021 10 01.
Article En | MEDLINE | ID: mdl-34364898

The anterior cingulate cortex (ACC) plays an important role in pain modulation, and pain-related emotional disorders. In the ACC, two major forms of long-term potentiation (LTP) coexist in excitatory synapses and lay the basis of chronic pain and pain-related emotional disorders. The induction of postsynaptic LTP is dependent on the activation of postsynaptic NMDA receptors (NMDARs), while the presynaptic LTP is NMDAR-independent. Long-term depression (LTD) can also be divided into two types according to the degree of sensitivity to the inhibition of NMDARs. NMDAR heteromers containing GluN2A and GluN2B act as key molecules in both the NMDAR-dependent postsynaptic LTP and LTD. Additionally, NMDARs also exist in presynaptic terminals and modulate the evoked and spontaneous transmitter release. From a translational point of view, inhibiting subtypes of NMDARs and/or downstream signaling proteins may provide potential drug targets for chronic pain and its related emotional disorders. This article is part of the special Issue on 'Glutamate Receptors -NMDA receptors'.


Gyrus Cinguli/physiology , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Animals , Gyrus Cinguli/drug effects , Humans , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression , Neuronal Plasticity/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Synapses/drug effects
13.
J Neurosci ; 41(38): 8075-8087, 2021 09 22.
Article En | MEDLINE | ID: mdl-34380767

Despite many observations of anterior cingulate cortex (ACC) activity related to cognition and affect in humans and nonhuman animals, little is known about the causal role of the ACC in psychological processes. Here, we investigate the causal role of the ACC in affective responding to threat in rhesus monkeys (Macaca mulatta), a species with an ACC largely homologous to humans in structure and connectivity. Male adult monkeys received bilateral ibotenate axon-sparing lesions to the ACC (sulcus and gyrus of areas 24, 32, and 25) and were tested in two classic tasks of monkey threat processing: the human intruder and object responsiveness tasks. Monkeys with ACC lesions did not significantly differ from controls in their overall mean reactivity toward threatening or novel stimuli. However, while control monkeys maintained their reactivity across test days, monkeys with ACC lesions reduced their reactivity toward stimuli as days advanced. Critically, this attenuated reactivity was found even when the stimuli presented each day were novel, suggesting that ACC lesions did not simply cause accelerated adaptation to stimuli as they became less novel over repeated presentations. Rather, these results imply that the primate ACC is necessary for maintaining appropriate affective responses toward potentially harmful and/or novel stimuli. These findings therefore have implications for mood disorders in which responding to threat and novelty is disrupted.SIGNIFICANCE STATEMENT Decades of research in humans and nonhuman animals have investigated the role of the anterior cingulate cortex in a huge number and variety of psychological processes spanning cognition and affect, as well as in psychological and neurologic diseases. The structure is broadly implicated in psychological processes and mental and neurologic health, yet its causal role in these processes has largely gone untested, particularly in primates. Here we demonstrate that when anterior cingulate cortex is completely eliminated, rhesus monkeys are initially responsive to threats, but these responses attenuate rather than persist, resembling a pattern of behavior commonly seen in patients diagnosed with mood disorders.


Affect/physiology , Attention/physiology , Cognition/physiology , Gyrus Cinguli/physiology , Neurons/physiology , Animals , Attention/drug effects , Cognition/drug effects , Gyrus Cinguli/drug effects , Ibotenic Acid , Macaca mulatta , Magnetic Resonance Imaging , Male , Neurons/drug effects
14.
Am J Psychiatry ; 178(9): 829-837, 2021 09 01.
Article En | MEDLINE | ID: mdl-34256607

OBJECTIVE: Although gabapentin has demonstrated efficacy in mitigating alcohol withdrawal symptoms and preventing relapse drinking in individuals with alcohol use disorder (AUD), the neurobiological mechanisms of action underlying these therapeutic effects remain unknown. The present study evaluated changes in GABA and glutamate levels in the dorsal anterior cingulate cortex (dACC) as candidate mechanisms of action. METHODS: In a 16-week randomized clinical trial, 68 adults with AUD, including a history of alcohol withdrawal syndrome, received 1,200 mg/day of gabapentin (N=37) or placebo (N=31) and nine medical management visits after ≥72 hours of abstinence. Proton MR spectroscopy (1H-MRS) estimates of dACC levels of GABA (N=67) and glutamate (N=64) were acquired before start of treatment and again approximately 14 days after randomization. Percent days abstinent was reported via timeline followback interview. RESULTS: The effects of gabapentin on GABA and glutamate levels were significantly associated with participants' percent days abstinent during early treatment. Specifically, gabapentin was associated with greater increases in glutamate and greater decreases in GABA levels in participants who remained mostly or entirely abstinent, and yet the opposite in participants who drank on more than half of the days preceding the second scan. Furthermore, gabapentin-treated participants with greater increases in glutamate levels during early treatment had significantly more percent days abstinent across the remainder of the study, relative to placebo-treated participants. CONCLUSIONS: In addition to providing insight into the mechanisms through which gabapentin may promote abstinence in individuals with AUD, this study also provides evidence for a biomarker of efficacious treatment that may be used to evaluate other glutamatergic or GABAergic medications for AUD and related conditions.


Alcoholism/drug therapy , Gabapentin/therapeutic use , Glutamic Acid/metabolism , Gyrus Cinguli/drug effects , gamma-Aminobutyric Acid/metabolism , Adult , Double-Blind Method , Female , Gabapentin/pharmacology , Gyrus Cinguli/metabolism , Humans , Male , Middle Aged , Proton Magnetic Resonance Spectroscopy , Substance Withdrawal Syndrome/drug therapy
15.
J Neurosci ; 41(35): 7461-7478, 2021 09 01.
Article En | MEDLINE | ID: mdl-34315810

Temporal costs influence reward-based decisions. This is commonly studied in temporal discounting tasks that involve choosing between cues signaling an imminent reward option or a delayed reward option. However, it is unclear whether the temporal delay before a reward can alter the value of that option. To address this, we identified the relative preference between different flavored rewards during a free-feeding test using male and female rats. Animals underwent training where either the initial preferred or the initial less preferred reward was delivered noncontingently. By manipulating the intertrial interval during training sessions, we could determine whether temporal delays impact reward preference in a subsequent free-feeding test. Rats maintained their initial preference if the same delays were used across all training sessions. When the initial less preferred option was delivered after short delays (high reward rate) and the initial preferred option was delivered after long delays (low reward rate), rats expectedly increased their preference for the initial less desirable option. However, rats also increased their preference for the initial less desirable option under the opposite training contingencies: delivering the initial less preferred reward after long delays and the initial preferred reward after short delays. These data suggest that sunk temporal costs enhance the preference for a less desirable reward option. Pharmacological and lesion experiments were performed to identify the neural systems responsible for this behavioral phenomenon. Our findings demonstrate the basolateral amygdala and retrosplenial cortex are required for temporal delays to enhance the preference for an initially less desirable reward.SIGNIFICANCE STATEMENT The goal of this study was to determine how temporal delays influence reward preference. We demonstrate that delivering an initially less desirable reward after long delays subsequently increases the consumption and preference for that reward. Furthermore, we identified the basolateral amygdala and the retrosplenial cortex as essential nuclei for mediating the change in reward preference elicited by sunk temporal costs.


Basolateral Nuclear Complex/physiology , Choice Behavior/physiology , Gyrus Cinguli/physiology , Reward , Time Factors , Animals , Feeding Behavior/drug effects , Feeding Behavior/physiology , Female , Flupenthixol/pharmacology , Food Preferences , Gyrus Cinguli/drug effects , Male , N-Methylaspartate/toxicity , Rats , Rats, Sprague-Dawley
16.
Neurosci Lett ; 762: 136146, 2021 09 25.
Article En | MEDLINE | ID: mdl-34332028

INTRODUCTION: Current treatments for cocaine use disorder (CUD) are not very effective and better treatments are needed. This study investigates the effectiveness of a combined intervention that targets the assumed underlying glutamate pathology in cocaine users. To this end, the combined effects of N-acetylcysteine (NAC) and working memory (WM) training on glutamate concentrations in the dorsal and rostral ACC were investigated in a randomized, double-blind placebo-controlled design. METHODS: In this study, 38 regular cocaine-using men were randomized to either 25-days with 2400 mg/day NAC and WM-training or 25 days with placebo with WM-training. Cocaine use, impulsivity, and glutamate concentrations in the dACC and rACC using proton Magnetic Resonance Spectroscopy were assessed at baseline and after treatment. RESULTS: Twenty-four participants completed the study, of which 9 received NAC and 15 received placebo. There were no baseline correlations of glutamate concentrations in the dACC or rACC with cocaine use measures or impulsivity. Additionally, there were no effects of NAC, WM-training, or the combination thereof on (changes in) glutamate concentrations in the dACC or rACC. DISCUSSION: This randomized proof of concept study could not confirm our hypotheses. Possible explanations are insufficient power and the possible absence of deviant baseline glutamate concentrations in the included participants. Future studies should consider larger samples and a non-using control group to confirm baseline deviations in glutamate in cocaine users.


Acetylcysteine/therapeutic use , Cocaine-Related Disorders/therapy , Glutamic Acid/drug effects , Gyrus Cinguli/drug effects , Learning , Adult , Double-Blind Method , Glutamic Acid/metabolism , Gyrus Cinguli/metabolism , Humans , Male , Memory, Short-Term , Middle Aged , Proof of Concept Study
17.
Neurosci Lett ; 761: 136119, 2021 09 14.
Article En | MEDLINE | ID: mdl-34280506

Complex Regional Pain Syndrome (CRPS) is a musculoskeletal pain condition that often develops after limb injury and/or immobilization. Although the exact mechanisms underlying CRPS are unknown, the syndrome is associated with central and autonomic nervous system dysregulation and peripheral hyperalgesia symptoms. These symptoms also manifest in alcoholic neuropathy, suggesting that the two conditions may be pathophysiologically accretive. Interestingly, people assigned female at birth (AFAB) appear to be more sensitive to both CRPS and alcoholic neuropathy. To better understand the biobehavioral mechanisms underlying these conditions, we investigated a model of combined CRPS and alcoholic neuropathy in female rats. Animals were pair-fed either a Lieber-DeCarli alcohol liquid diet or a control diet for ten weeks. CRPS was modeled via unilateral hind limb cast immobilization for seven days, allowing for the other limb to serve as a within-subject control for hyperalgesia measures. To investigate the role of circulating ovarian hormones on pain-related behaviors, half of the animals underwent ovariectomy (OVX). Using the von Frey procedure to record mechanical paw withdrawal thresholds, we found that cast immobilization and chronic alcohol drinking separately and additively produced mechanical hyperalgesia observed 3 days after cast removal. We then examined neuroadaptations in AMPA GluR1 and NMDA NR1 glutamate channel subunits, extracellular signal-regulated kinase (ERK), and cAMP response element-binding protein (CREB) in bilateral motor and cingulate cortex across all groups. Consistent with increased pain-related behavior, chronic alcohol drinking increased GluR1, NR1, ERK, and CREB phosphorylation in the cingulate cortex. OVX did not alter any of the observed effects. Our results suggest accretive relationships between CRPS and alcoholic neuropathy symptoms and point to novel therapeutic targets for these conditions.


Alcohol Drinking/metabolism , Gyrus Cinguli/drug effects , Hyperalgesia/physiopathology , MAP Kinase Signaling System , Alcohol Drinking/physiopathology , Animals , Central Nervous System Stimulants/toxicity , Cyclic AMP Response Element-Binding Protein/metabolism , Ethanol/pharmacology , Ethanol/toxicity , Female , Gyrus Cinguli/metabolism , Hindlimb Suspension/adverse effects , Hyperalgesia/etiology , Hyperalgesia/metabolism , Motor Cortex/drug effects , Motor Cortex/metabolism , Nociception , Rats , Rats, Inbred F344 , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
18.
Cell Rep ; 36(3): 109411, 2021 07 20.
Article En | MEDLINE | ID: mdl-34289348

Oxytocin is a well-known neurohypophysial hormone that plays an important role in behavioral anxiety and nociception. Two major forms of long-term potentiation, presynaptic LTP (pre-LTP) and postsynaptic LTP (post-LTP), have been characterized in the anterior cingulate cortex (ACC). Both pre-LTP and post-LTP contribute to chronic-pain-related anxiety and behavioral sensitization. The roles of oxytocin in the ACC have not been studied. Here, we find that microinjections of oxytocin into the ACC attenuate nociceptive responses and anxiety-like behavioral responses in animals with neuropathic pain. Application of oxytocin selectively blocks the maintenance of pre-LTP but not post-LTP. In addition, oxytocin enhances inhibitory transmission and excites ACC interneurons. Similar results are obtained by using selective optical stimulation of oxytocin-containing projecting terminals in the ACC in animals with neuropathic pain. Our results demonstrate that oxytocin acts on central synapses and reduces chronic-pain-induced anxiety by reducing pre-LTP.


Anxiety/physiopathology , Emotions , Gyrus Cinguli/pathology , Long-Term Potentiation , Neuralgia/pathology , Neuralgia/physiopathology , Oxytocin/pharmacology , Presynaptic Terminals/pathology , Analgesics/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Calcium/metabolism , Chronic Pain/pathology , Chronic Pain/physiopathology , Emotions/drug effects , Female , Gyrus Cinguli/drug effects , Gyrus Cinguli/physiopathology , Interneurons/drug effects , Light , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Microinjections , Nerve Tissue/drug effects , Nerve Tissue/pathology , Nerve Tissue/physiopathology , Neural Inhibition/drug effects , Neuralgia/complications , Oxytocin/administration & dosage , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/pathology , Paraventricular Hypothalamic Nucleus/physiopathology , Presynaptic Terminals/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, GABA-A/metabolism , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Up-Regulation/drug effects
19.
Commun Biol ; 4(1): 662, 2021 06 02.
Article En | MEDLINE | ID: mdl-34079054

Pathological impulsivity is a debilitating symptom of multiple psychiatric diseases with few effective treatment options. To identify druggable receptors with anti-impulsive action we developed a systematic target discovery approach combining behavioural chemogenetics and gene expression analysis. Spatially restricted inhibition of three subdivisions of the prefrontal cortex of mice revealed that the anterior cingulate cortex (ACC) regulates premature responding, a form of motor impulsivity. Probing three G-protein cascades with designer receptors, we found that the activation of Gi-signalling in layer-5 pyramidal cells (L5-PCs) of the ACC strongly, reproducibly, and selectively decreased challenge-induced impulsivity. Differential gene expression analysis across murine ACC cell-types and 402 GPCRs revealed that - among Gi-coupled receptor-encoding genes - Grm2 is the most selectively expressed in L5-PCs while alternative targets were scarce. Validating our approach, we confirmed that mGluR2 activation reduced premature responding. These results suggest Gi-coupled receptors in ACC L5-PCs as therapeutic targets for impulse control disorders.


GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Gyrus Cinguli/cytology , Gyrus Cinguli/physiology , Pyramidal Cells/physiology , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Female , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Expression/drug effects , Gyrus Cinguli/drug effects , Humans , Impulsive Behavior/drug effects , Impulsive Behavior/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/physiology , Signal Transduction
20.
Addict Biol ; 26(6): e13048, 2021 11.
Article En | MEDLINE | ID: mdl-33973711

The process through which early memories are transferred to the cerebral cortex to form long-term memories is referred to as memory consolidation, and the basolateral amygdala (BLA) is an important brain region involved in this process. Although functional connections between the BLA and multiple brain regions are critical for the consolidation of withdrawal memory, whether the projection from the BLA to the anterior cingulate cortex (ACC) is involved in the formation or consolidation of withdrawal memory remains unclear. In this paper, we used a chemical genetic method to specifically label the BLA-ACC projection in a combined morphine withdrawal and conditioned place aversion (CPA) animal model. We found that (1) the inhibition of the BLA-ACC projection during conditioning had no effects on the formation of early withdrawal memory; (2) the inhibition of the BLA-ACC projection had no effects on the retrieval of either early or long-term withdrawal memory; and (3) the persistent inhibition of the BLA-ACC projection after early withdrawal memory formation could inhibit the formation of long-term withdrawal memory and decrease Arc protein expression in the ACC. These results suggested that the persistent activation of the BLA-ACC projection after the formation of early withdrawal memory facilitates the formation of long-term withdrawal memory by increasing the plasticity of ACC neurons.


Basolateral Nuclear Complex/drug effects , Gyrus Cinguli/drug effects , Memory Consolidation/drug effects , Morphine/pharmacology , Narcotic-Related Disorders/physiopathology , Animals , Gyrus Cinguli/metabolism , Male , Memory Consolidation/physiology , Memory, Long-Term/physiology , Mice , Mice, Inbred C57BL
...