Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cell ; 159(1): 33-45, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25259918

ABSTRACT

Depression is a debilitating condition with a profound impact on quality of life for millions of people worldwide. Physical exercise is used as a treatment strategy for many patients, but the mechanisms that underlie its beneficial effects remain unknown. Here, we describe a mechanism by which skeletal muscle PGC-1α1 induced by exercise training changes kynurenine metabolism and protects from stress-induced depression. Activation of the PGC-1α1-PPARα/δ pathway increases skeletal muscle expression of kynurenine aminotransferases, thus enhancing the conversion of kynurenine into kynurenic acid, a metabolite unable to cross the blood-brain barrier. Reducing plasma kynurenine protects the brain from stress-induced changes associated with depression and renders skeletal muscle-specific PGC-1α1 transgenic mice resistant to depression induced by chronic mild stress or direct kynurenine administration. This study opens therapeutic avenues for the treatment of depression by targeting the PGC-1α1-PPAR axis in skeletal muscle, without the need to cross the blood-brain barrier.


Subject(s)
Depression/prevention & control , Kynurenine/metabolism , Muscle, Skeletal/enzymology , Stress, Psychological/complications , Transcription Factors/metabolism , Animals , Blood-Brain Barrier , Depression/metabolism , Gene Expression Profiling , Humans , Kynurenic Acid , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Conditioning, Animal , Physical Conditioning, Human , Transaminases/metabolism , Transcription Factors/genetics
2.
J Neuroinflammation ; 20(1): 3, 2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36593462

ABSTRACT

BACKGROUND: Astrocytes are crucial for maintaining brain homeostasis and synaptic function, but are also tightly connected to the pathogenesis of Alzheimer's disease (AD). Our previous data demonstrate that astrocytes ingest large amounts of aggregated amyloid-beta (Aß), but then store, rather than degrade the ingested material, which leads to severe cellular stress. However, the involvement of pathological astrocytes in AD-related synaptic dysfunction remains to be elucidated. METHODS: In this study, we aimed to investigate how intracellular deposits of Aß in astrocytes affect their interplay with neurons, focusing on neuronal function and viability. For this purpose, human induced pluripotent stem cell (hiPSC)-derived astrocytes were exposed to sonicated Αß42 fibrils. The direct and indirect effects of the Αß-exposed astrocytes on hiPSC-derived neurons were analyzed by performing astrocyte-neuron co-cultures as well as additions of conditioned media or extracellular vesicles to pure neuronal cultures. RESULTS: Electrophysiological recordings revealed significantly decreased frequency of excitatory post-synaptic currents in neurons co-cultured with Aß-exposed astrocytes, while conditioned media from Aß-exposed astrocytes had the opposite effect and resulted in hyperactivation of the synapses. Clearly, factors secreted from control, but not from Aß-exposed astrocytes, benefited the wellbeing of neuronal cultures. Moreover, reactive astrocytes with Aß deposits led to an elevated clearance of dead cells in the co-cultures. CONCLUSIONS: Taken together, our results demonstrate that inclusions of aggregated Aß affect the reactive state of the astrocytes, as well as their ability to support neuronal function.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Humans , Astrocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Culture Media, Conditioned/pharmacology , Cells, Cultured , Amyloid beta-Peptides/pharmacology , Amyloid beta-Peptides/metabolism , Neurons/metabolism , Alzheimer Disease/pathology
3.
J Neuroinflammation ; 20(1): 43, 2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36803838

ABSTRACT

BACKGROUND: Astrocytes play a central role in maintaining brain energy metabolism, but are also tightly connected to the pathogenesis of Alzheimer's disease (AD). Our previous studies demonstrate that inflammatory astrocytes accumulate large amounts of aggregated amyloid-beta (Aß). However, in which way these Aß deposits influence their energy production remain unclear. METHODS: The aim of the present study was to investigate how Aß pathology in astrocytes affects their mitochondria functionality and overall energy metabolism. For this purpose, human induced pluripotent cell (hiPSC)-derived astrocytes were exposed to sonicated Aß42 fibrils for 7 days and analyzed over time using different experimental approaches. RESULTS: Our results show that to maintain stable energy production, the astrocytes initially increased their mitochondrial fusion, but eventually the Aß-mediated stress led to abnormal mitochondrial swelling and excessive fission. Moreover, we detected increased levels of phosphorylated DRP-1 in the Aß-exposed astrocytes, which co-localized with lipid droplets. Analysis of ATP levels, when blocking certain stages of the energy pathways, indicated a metabolic shift to peroxisomal-based fatty acid ß-oxidation and glycolysis. CONCLUSIONS: Taken together, our data conclude that Aß pathology profoundly affects human astrocytes and changes their entire energy metabolism, which could result in disturbed brain homeostasis and aggravated disease progression.


Subject(s)
Alzheimer Disease , Astrocytes , Humans , Astrocytes/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Alzheimer Disease/pathology , Energy Metabolism , Mitochondria/pathology
4.
J Biol Chem ; 297(6): 101355, 2021 12.
Article in English | MEDLINE | ID: mdl-34717959

ABSTRACT

The ion pump Na+,K+-ATPase is a critical determinant of neuronal excitability; however, its role in the etiology of diseases of the central nervous system (CNS) is largely unknown. We describe here the molecular phenotype of a Trp931Arg mutation of the Na+,K+-ATPase catalytic α1 subunit in an infant diagnosed with therapy-resistant lethal epilepsy. In addition to the pathological CNS phenotype, we also detected renal wasting of Mg2+. We found that membrane expression of the mutant α1 protein was low, and ion pumping activity was lost. Arginine insertion into membrane proteins can generate water-filled pores in the plasma membrane, and our molecular dynamic (MD) simulations of the principle states of Na+,K+-ATPase transport demonstrated massive water inflow into mutant α1 and destabilization of the ion-binding sites. MD simulations also indicated that a water pathway was created between the mutant arginine residue and the cytoplasm, and analysis of oocytes expressing mutant α1 detected a nonspecific cation current. Finally, neurons expressing mutant α1 were observed to be depolarized compared with neurons expressing wild-type protein, compatible with a lowered threshold for epileptic seizures. The results imply that Na+,K+-ATPase should be considered a neuronal locus minoris resistentia in diseases associated with epilepsy and with loss of plasma membrane integrity.


Subject(s)
Epilepsy/genetics , Mutation, Missense , Sodium-Potassium-Exchanging ATPase/genetics , Animals , Anticonvulsants/pharmacology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cells, Cultured , Drug Resistance , Epilepsy/drug therapy , Epilepsy/pathology , Humans , Infant , Molecular Dynamics Simulation , Mutation, Missense/drug effects , Protein Subunits/analysis , Protein Subunits/genetics , Sodium-Potassium-Exchanging ATPase/analysis , Xenopus
5.
Int J Mol Sci ; 22(4)2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33672070

ABSTRACT

Serotonin communication operates mainly in the extracellular space and cerebrospinal fluid (CSF), using volume transmission with serotonin moving from source to target cells (neurons and astroglia) via energy gradients, leading to the diffusion and convection (flow) of serotonin. One emerging concept in depression is that disturbances in the integrative allosteric receptor-receptor interactions in highly vulnerable 5-HT1A heteroreceptor complexes can contribute to causing major depression and become novel targets for the treatment of major depression (MD) and anxiety. For instance, a disruption and/or dysfunction in the 5-HT1A-FGFR1 heteroreceptor complexes in the raphe-hippocampal serotonin neuron systems can contribute to the development of MD. It leads inter alia to reduced neuroplasticity and potential atrophy in the raphe-cortical and raphe-striatal 5-HT pathways and in all its forebrain networks. Reduced 5-HT1A auto-receptor function, increased plasticity and trophic activity in the midbrain raphe 5-HT neurons can develop via agonist activation of allosteric receptor-receptor interactions in the 5-HT1A-FGFR1 heterocomplex. Additionally, the inhibitory allosteric receptor-receptor interactions in the 5-HT1AR-5-HT2AR isoreceptor complex therefore likely have a significant role in modulating mood, involving a reduction of postjunctional 5-HT1AR protomer signaling in the forebrain upon activation of the 5-HT2AR protomer. In addition, oxytocin receptors (OXTRs) play a significant and impressive role in modulating social and cognitive related behaviors like bonding and attachment, reward and motivation. Pathological blunting of the OXTR protomers in 5-HT2AR and especially in 5-HT2CR heteroreceptor complexes can contribute to the development of depression and other types of psychiatric diseases involving disturbances in social behaviors. The 5-HTR heterocomplexes are novel targets for the treatment of MD.


Subject(s)
Depression/metabolism , Depressive Disorder, Major/metabolism , Hippocampus/metabolism , Neurons/metabolism , Serotonin/metabolism , Signal Transduction , Animals , Humans , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Receptors, Oxytocin/metabolism
6.
Int J Neuropsychopharmacol ; 21(7): 668-676, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29688411

ABSTRACT

The diagnosis of a mental disorder generally depends on clinical observations and phenomenological symptoms reported by the patient. The definition of a given diagnosis is criteria based and relies on the ability to accurately interpret subjective symptoms and complex behavior. This type of diagnosis comprises a challenge to translate to reliable animal models, and these translational uncertainties hamper the development of new treatments. In this review, we will discuss how depressive-like behavior can be induced in rodents, and the relationship between these models and depression in humans. Specifically, we suggest similarities between triggers of depressive-like behavior in animal models and human conditions known to increase the risk of depression, for example exhaustion and bullying. Although we acknowledge the potential problems in comparing animal findings to human conditions, such comparisons are useful for understanding the complexity of depression, and we highlight the need to develop clinical diagnoses and animal models in parallel to overcome translational uncertainties.


Subject(s)
Behavior, Animal/physiology , Depressive Disorder , Disease Models, Animal , Neuropsychiatry/standards , Rodentia , Translational Research, Biomedical/standards , Animals , Depressive Disorder/etiology , Depressive Disorder/genetics , Depressive Disorder/immunology , Depressive Disorder/physiopathology , Mice , Rats
7.
Int J Neuropsychopharmacol ; 18(9)2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25762718

ABSTRACT

BACKGROUND: Histamine is a modulatory neurotransmitter regulating neuronal activity. Antidepressant drugs target modulatory neurotransmitters, thus ultimately regulating glutamatergic transmission and plasticity. Histamine H3 receptor (H3R) antagonists have both pro-cognitive and antidepressant effects; however, the mechanism by which they modulate glutamate transmission is not clear. We measured the effects of the H3R antagonist clobenpropit in the Flinders Sensitive Line (FSL), a rat model of depression with impaired memory and altered glutamatergic transmission. METHODS: Behavioral tests included the forced swim test, memory tasks (passive avoidance, novel object recognition tests), and anxiety-related paradigms (novelty suppressed feeding, social interaction, light/dark box tests). Hippocampal protein levels were detected by Western blot. Hippocampal plasticity was studied by in slice field recording of CA3-CA1 long-term synaptic potentiation (LTP), and glutamatergic transmission by whole-cell patch clamp recording of excitatory postsynaptic currents (EPSCs) in CA1 pyramidal neurons. RESULTS: Clobenpropit, administered systemically or directly into the hippocampus, decreased immobility during the forced swim test; systemic injections reversed memory deficits and increased hippocampal GluN2A protein levels. FSL rats displayed anxiety-related behaviors not affected by clobenpropit treatment. Clobenpropit enhanced hippocampal plasticity, but did not affect EPSCs. H1R and H2R antagonists prevented the clobenpropit-induced increase in LTP and, injected locally into the hippocampus, blocked clobenpropit's effect in the forced swim test. CONCLUSIONS: Clobenpropit's antidepressant effects and the enhanced synaptic plasticity require hippocampal H1R and H2R activation, suggesting that clobenpropit acts through disinhibition of histamine release. Clobenpropit reverses memory deficits and increases hippocampal GluN2A expression without modifying anxiety-related phenotypes or EPSCs in CA1 pyramidal neurons.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Hippocampus/drug effects , Histamine H3 Antagonists/pharmacology , Imidazoles/pharmacology , Long-Term Potentiation/drug effects , Thiourea/analogs & derivatives , Animals , Antidepressive Agents/administration & dosage , Anxiety/drug therapy , Behavior, Animal/drug effects , Disease Models, Animal , Histamine H3 Antagonists/administration & dosage , Imidazoles/administration & dosage , Male , Memory Disorders/drug therapy , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Rats , Rats, Sprague-Dawley , Thiourea/administration & dosage , Thiourea/pharmacology
8.
BMC Neurosci ; 15: 45, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24670218

ABSTRACT

BACKGROUND: Norbin is a neuron-specific, cytosolic protein that interacts with the metabotropic glutamate receptor 5 (mGluR5) and has a profound impact on mGluR5 signaling. Yet, little is known about its synaptic distribution. RESULTS: Here we have analyzed the spatial relationship between Norbin, postsynaptic density protein 95 (PSD-95), actin and mGluR5 in spines using super-resolution microscopy. Norbin was found to have a high degree of colocalization with actin and a lower degree of colocalization with PSD-95. Co-immunoprecipitation studies confirmed that interaction occurs between Norbin and actin, but not between Norbin and PSD-95. Norbin was also found to have a high degree of colocalization with the perisynaptically located mGluR5. Findings based on structured illumination microscopy (3D-SIM) of exogenous expressed Norbin-GFP were confirmed by stimulated emission depletion microscopy (STED) of immunolabeled endogenous Norbin. CONCLUSIONS: Norbin associates with actin rather than with PSD-95 in dendritic spines. Results regarding protein localization and colocalization performed with conventional confocal microscopy must be interpreted with great caution. The now available super-resolution microscopy techniques provide more accurate information about sub-cellular protein localization than previously was possible.


Subject(s)
Actins/metabolism , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Neuropeptides/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Subcellular Fractions/metabolism , Animals , Cells, Cultured , Female , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
9.
FASEB J ; 27(9): 3564-71, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23709617

ABSTRACT

Hospitalization for major surgery or critical illness often associates with cognitive decline. Inflammation and dysregulation of the innate immune system can exert broad effects in the periphery and central nervous system (CNS), yet the mechanisms underlying memory impairment after surgery remain poorly understood and without effective therapy. Endogenous regulation of acute inflammation is providing novel approaches to treat several disease states including sepsis, pain, obesity and diabetes. Resolvins are potent endogenous lipid mediators biosynthesized during the resolution phase of acute inflammation that display immunoresolvent actions. Here, using a mouse model of surgery-induced cognitive decline we report that orthopedic surgery affects hippocampal neuronal-glial function, including synaptic transmission and plasticity. Systemic prophylaxis with aspirin-triggered resolvin D1 (AT-RvD1: 7S,8R,17R-trihydroxy-4Z,9E,11E,13Z,15E,19Z-docosahexaenoic acid, as little as 100 ng dose per mouse) improved memory decline following surgery and abolished signs of synaptic dysfunction. Moreover, delayed administration 24 h after surgery also attenuated signs of neuronal dysfunction postoperatively. AT-RvD1 also limited peripheral damage by modulating the release of systemic interleukin (IL)-6 and improved other clinical markers of tissue injury. Collectively, these results demonstrate a novel role of AT-RvD1 in modulating the proinflammatory milieu after aseptic injury and protecting the brain from neuroinflammation, synaptic dysfunction and cognitive decline. These findings provide novel and safer approaches to treat postoperative cognitive decline and potentially other forms of memory dysfunctions.


Subject(s)
Aspirin/pharmacology , Aspirin/therapeutic use , Cognition/drug effects , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Electrophysiology , Immunohistochemistry , In Vitro Techniques , Locomotion/drug effects , Long-Term Potentiation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Postoperative Complications/prevention & control , Superoxides/metabolism
10.
Proc Natl Acad Sci U S A ; 108(2): 828-33, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187407

ABSTRACT

Prolonged AMPA-receptor blockade in hippocampal neuron cultures leads to both an increased expression of GluA1 postsynaptically and an increase in vesicle pool size and turnover rate presynaptically, adaptive changes that extend beyond simple synaptic scaling. As a molecular correlate, expression of the ß Ca(2+)/CaM-dependent kinase type II (ßCaMKII) is increased in response to synaptic inactivity. Here we set out to clarify the role of ßCaMKII in the various manifestations of adaptation. Knockdown of ßCaMKII by lentiviral-mediated expression of shRNA prevented the synaptic inactivity-induced increase in GluA1, as did treatment with the CaM kinase inhibitor KN-93, but not the inactive analog KN-92. These results demonstrate that, spurred by AMPA-receptor blockade, up-regulation of ßCaMKII promotes increased GluA1 expression. Indeed, transfection of ßCaMKII, but not a kinase-dead mutant, increased GluA1 expression on dendrites and elevated vesicle turnover (Syt-Ab uptake), mimicking the effect of synaptic inactivity on both sides of the synapse. In cells with elevated ßCaMKII, relief of synaptic-activity blockade uncovered an increase in the frequency of miniature excitatory postsynaptic currents that could be rapidly and fully suppressed by PhTx blockade of GluA1 receptors. This increased mini frequency involved a genuine presynaptic enhancement, not merely an increased abundance of synapses. This finding suggests that Ca(2+) flux through GluA1 receptors may trigger the acute release of a retrograde messenger. Taken together, our results indicate that synaptic inactivity-induced increases in ßCaMKII expression set in motion a series of events that culminate in coordinated pre- and postsynaptic adaptations in synaptic transmission.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Synaptic Transmission , Animals , Benzylamines/pharmacology , Green Fluorescent Proteins/metabolism , Homeostasis , Lentivirus/genetics , Mice , Microscopy, Fluorescence/methods , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction , Sulfonamides/pharmacology , Up-Regulation
11.
J Alzheimers Dis ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38995780

ABSTRACT

Background: Alzheimer's disease (AD) is the most common neurodegenerative disease. Unfortunately, efficient and affordable treatments are still lacking for this neurodegenerative disorder, it is therefore urgent to identify new pharmacological targets. Astrocytes are playing a crucial role in the tuning of synaptic transmission and several studies have pointed out severe astrocyte reactivity in AD. Reactive astrocytes show altered physiology and function, suggesting they could have a role in the early pathophysiology of AD. Objective: We aimed to characterize early synaptic impairments in the AppNL-F knock-in mouse model of AD, especially to understand the contribution of astrocytes to early brain dysfunctions. Methods: The AppNL-F mouse model carries two disease-causing mutations inserted in the amyloid precursor protein gene. This strain does not start to develop amyloid-ß plaques until 9 months of age. Thanks to electrophysiology, we investigated synaptic function, at both neuronal and astrocytic levels, in 6-month-old animals and correlate the synaptic activity with emotional behavior. Results: Electrophysiological recordings in the hippocampus revealed an overall synaptic mistuning at a pre-plaque stage of the pathology, associated to an intact social memory but a stronger depressive-like behavior. Astrocytes displayed a reactive-like morphology and a higher tonic GABA current compared to control mice. Interestingly, we here show that the synaptic impairments in hippocampal slices are partially corrected by a pre-treatment with the monoamine oxidase B blocker deprenyl or the fast-acting antidepressant ketamine (5 mg/kg). Conclusions: We propose that reactive astrocytes can induce synaptic mistuning early in AD, before plaques deposition, and that these changes are associated with emotional symptoms.

12.
Int J Neuropsychopharmacol ; 16(9): 2041-51, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23672849

ABSTRACT

Dopamine modulates cognitive functions through regulation of synaptic transmission and plasticity in the hippocampus and prefrontal cortex (PFC). Thus, dopamine dysfunction in depression may be particularly relevant for the cognitive symptoms. The norepinephrine transporter inhibitor reboxetine facilitates memory processing in both healthy volunteers and in depressed patients and increases dopamine release in both the hippocampus and PFC. We investigated the potential involvement of the hippocampal and PFC dopamine D1/5 receptors in the cognitive effects of reboxetine using the object recognition test in rats. Infusion of the D1/5 antagonist SCH23390 into the dorsal hippocampus or medial PFC prior to the exploration of the objects impaired memory. Conversely, infusion of the D1/5 agonist SKF81297 into the dorsal hippocampus or medial PFC facilitated memory. Reboxetine similarly facilitated recognition memory in healthy rats and the D1/5 antagonist SCH23390 reversed this effect when infused into the dorsal PFC, but not when infused into the hippocampus. Moreover, systemic reboxetine increased the levels of the NMDA subunit GluN2A in the PFC but not in the hippocampus. Finally, we demonstrate that a single dose of reboxetine does not affect immobility in the forced swim test but improves recognition memory in the Flinders sensitive line (FSL) rat model for depression. The present data in rats are in line with effects of reboxetine on memory formation in healthy volunteers and depressed patients and indicate the involvement of PFC dopamine D1/5 receptors.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Depression/drug therapy , Hippocampus/drug effects , Morpholines/pharmacology , Nootropic Agents/pharmacology , Prefrontal Cortex/drug effects , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D5/drug effects , Recognition, Psychology/drug effects , Animals , Cognition/drug effects , Depression/metabolism , Depression/physiopathology , Depression/psychology , Disease Models, Animal , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Motor Activity/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Reboxetine , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D5/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Swimming , Time Factors
13.
Proc Natl Acad Sci U S A ; 107(50): 21806-11, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21098665

ABSTRACT

Prolonged blockade of AMPA-type glutamate receptors in hippocampal neuron cultures leads to homeostatic enhancements of pre- and postsynaptic function that appear correlated at individual synapses, suggesting some form of transsynaptic coordination. The respective modifications are important for overall synaptic strength but their interrelationship, dynamics, and molecular underpinnings are unclear. Here we demonstrate that adaptation begins postsynaptically but is ultimately communicated to presynaptic terminals and expressed as an accelerated turnover of synaptic vesicles. Critical postsynaptic modifications occur over hours, but enable retrograde communication within minutes once AMPA receptor (AMPAR) blockade is removed, causing elevation of both spontaneous and evoked vesicle fusion. The retrograde signaling does not require spiking activity and can be interrupted by NBQX, philanthotoxin, postsynaptic BAPTA, or external sequestration of BDNF, consistent with the acute release of retrograde messenger, triggered by postsynaptic Ca(2+) elevation via Ca(2+)-permeable AMPARs.


Subject(s)
Homeostasis/physiology , Neurons/metabolism , Presynaptic Terminals/metabolism , Receptors, AMPA/antagonists & inhibitors , Synapses/metabolism , Action Potentials/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Neurons/cytology , Nitric Oxide/metabolism , Patch-Clamp Techniques , Receptors, AMPA/metabolism , Signal Transduction/physiology , Synaptic Vesicles/metabolism
14.
Neuropsychopharmacology ; 48(13): 1859-1868, 2023 12.
Article in English | MEDLINE | ID: mdl-37301901

ABSTRACT

Mistuning of synaptic transmission has been proposed to underlie many psychiatric disorders, with decreased reuptake of the excitatory neurotransmitter glutamate as one contributing factor. Synaptic tuning occurs through several diverging and converging forms of plasticity. By recording evoked field postsynaptic potentials in the CA1 area in hippocampal slices, we found that inhibiting glutamate transporters using DL-TBOA causes retuning of synaptic transmission, resulting in a new steady state with reduced synaptic strength and a lower threshold for inducing long-term synaptic potentiation (LTP). Moreover, a similar reduced threshold for LTP was observed in a rat model of depression with decreased levels of glutamate transporters. Most importantly, we found that the antidepressant ketamine counteracts the effects of increased glutamate on the various steps involved in synaptic retuning. We, therefore, propose that ketamine's mechanism of action as an antidepressant is to restore adequate synaptic tuning.


Subject(s)
Ketamine , Humans , Rats , Animals , Ketamine/pharmacology , Hippocampus , Long-Term Potentiation/physiology , Synaptic Transmission , Antidepressive Agents/pharmacology , Glutamates , Synapses
15.
Psychopharmacology (Berl) ; 240(8): 1789-1804, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37394539

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is defined as a complex mental disorder which is characterized by a pervasive low mood and aversion to activity. Several types of neurotransmitter systems e.g. serotonergic, glutamatergic and noradrenergic systems have been suggested to play an important role in the origination of depression, but neurotrophins such as brain derived neurotrophic factor (BDNF) have also been implicated in the disease process. OBJECTIVES: The purpose of this study was to examine the effects of a newly developed class of molecules, characterized as positive allosteric modulators of neurotrophin/Trk receptor mediated signaling (Trk-PAM), on neurotransmitter release and depression-like behavior in vivo. METHODS: The effect of and possible interaction of neurotrophin/Trk signaling pathways with serotonergic and glutamatergic systems in the modulation of depression-related responses was studied using newly developed Trk-PAM compounds (ACD855, ACD856 and AC26845), as well as ketamine and fluoxetine in the forced swim test (FST) in rodents. Moreover, in vivo microdialysis in freely moving rats was used to assess changes in neurotransmitter levels in the rat. RESULTS: The results from the study show that several different compounds, which all potentiate Trk-receptor mediated signaling, display antidepressant-like activity in the FST. Moreover, the data also indicate that the effects of both fluoxetine and ketamine in the FST, both used in clinical practice, are mediated via BDNF/TrkB signaling, which could have implications for novel therapies in MDD. CONCLUSIONS: Trk-PAMs could provide an interesting avenue for the development of novel therapeutics in this area.


Subject(s)
Depressive Disorder, Major , Ketamine , Rats , Animals , Fluoxetine/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder, Major/drug therapy , Ketamine/pharmacology , Antidepressive Agents/pharmacology , Receptor, trkB/metabolism
16.
Acta Neuropathol Commun ; 11(1): 97, 2023 06 17.
Article in English | MEDLINE | ID: mdl-37330529

ABSTRACT

Tau deposits in astrocytes are frequently found in Alzheimer's disease (AD) and other tauopathies. Since astrocytes do not express tau, the inclusions have been suggested to be of neuronal origin. However, the mechanisms behind their appearance and their relevance for disease progression remain unknown. Here we demonstrate, using a battery of experimental techniques that human astrocytes serve as an intermediator, promoting cell-to-cell spreading of pathological tau. Human astrocytes engulf and process, but fail to fully degrade dead neurons with tau pathology, as well as synthetic tau fibrils and tau aggregates isolated from AD brain tissue. Instead, the pathogenic tau is spread to nearby cells via secretion and tunneling nanotube mediated transfer. By performing co-culture experiments we could show that tau-containing astrocytes induce tau pathology in healthy human neurons directly. Furthermore, our results from a FRET based seeding assay, demonstrated that the tau proteoforms secreted by astrocytes have an exceptional seeding capacity, compared to the original tau species engulfed by the cells. Taken together, our study establishes a central role for astrocytes in mediating tau pathology, which could be of relevance for identifying novel treatment targets for AD and other tauopathies.


Subject(s)
Alzheimer Disease , Tauopathies , Humans , tau Proteins/metabolism , Astrocytes/metabolism , Tauopathies/pathology , Alzheimer Disease/pathology , Neurons/metabolism , Brain/pathology
18.
Int J Neuropsychopharmacol ; 15(4): 531-40, 2012 May.
Article in English | MEDLINE | ID: mdl-21669025

ABSTRACT

Abnormal dopaminergic neurotransmission in the hippocampus may be involved in certain aspects of cognitive dysfunction. In the hippocampus, there is little, if any, expression of dopamine transporters (DAT), indicating that the mechanism for dopamine clearance differs from that in the striatum. Here, by means of in-vivo microdialysis in freely moving rats, we tested the hypothesis that the norepinephrine transporter (NET) is involved in dopamine clearance in the hippocampus. We found that systemic administration of the selective NET inhibitor reboxetine (3 mg/kg) and the psychostimulants amphetamine (0.5 mg/kg) and cocaine (10 mg/kg) increased hippocampal dopamine efflux. Local administration of reboxetine (300 µM) produced a large increase in hippocampal dopamine levels that could not be further enhanced by the addition of the NET/DAT inhibitor nomifensine (100 µM). Administration of the specific DAT inhibitor GBR12909 at a concentration (1 mM) that robustly increased dopamine in the nucleus accumbens had a comparably smaller effect in the hippocampus. In line with a minor role of DAT in the hippocampus, we detected very little DAT in this area using ligand binding with radiolabelled RTI-55. Moreover, in contrast to raclopride (100 µM), a dopamine D2-autoreceptor antagonist, local administration of the α2-adrenoceptor antagonist idazoxan (100 µM) increased hippocampal dopamine. Taken together, our data demonstrate an interaction between dopamine and norepinephrine systems in the hippocampus. It is proposed that this interaction originates from a shared uptake mechanism at the NET level.


Subject(s)
Dopamine/metabolism , Hippocampus/metabolism , Norepinephrine Plasma Membrane Transport Proteins/physiology , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Amphetamine/pharmacology , Animals , Cocaine/pharmacology , Dopamine Antagonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Drug Administration Routes , Drug Interactions , Hippocampus/drug effects , Idazoxan/pharmacology , Male , Microdialysis , Morpholines/pharmacology , Nomifensine/pharmacology , Raclopride/pharmacology , Rats , Rats, Wistar , Reboxetine , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Time Factors , Wakefulness
19.
J Alzheimers Dis ; 90(2): 565-583, 2022.
Article in English | MEDLINE | ID: mdl-36155507

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) research has relied on mouse models overexpressing human mutant A ßPP; however, newer generation knock-in models allow for physiological expression of amyloid-ß protein precursor (AßPP) containing familial AD mutations where murine AßPP is edited with a humanized amyloid-ß (Aß) sequence. The AppNL-F mouse model has shown substantial similarities to AD brains developing late onset cognitive impairment. OBJECTIVE: In this study, we aimed to characterize mature primary cortical neurons derived from homozygous AppNL-F embryos, especially to identify early mitochondrial alterations in this model. METHODS: Primary cultures of AppNL-F neurons kept in culture for 12-15 days were used to measure Aß levels, secretase activity, mitochondrial functions, mitochondrial-ER contacts, synaptic function, and cell death. RESULTS: We detected higher levels of Aß42 released from AppNL-F neurons as compared to wild-type neurons. AppNL-F neurons, also displayed an increased Aß42/Aß40 ratio, similar to adult AppNL-F mouse brain. Interestingly, we found an upregulation in mitochondrial oxygen consumption with concomitant downregulation in glycolytic reserve. Furthermore, AppNL-F neurons were more susceptible to cell death triggered by mitochondrial electron transport chain inhibition. Juxtaposition between ER and mitochondria was found to be substantially upregulated, which may account for upregulated mitochondrial-derived ATP production. However, anterograde mitochondrial movement was severely impaired in this model along with loss in synaptic vesicle protein and impairment in pre- and post-synaptic function. CONCLUSION: We show that widespread mitochondrial alterations can be detected in AppNL-F neurons in vitro, where amyloid plaque deposition does not occur, suggesting soluble and oligomeric Aß-species being responsible for these alterations.


Subject(s)
Alzheimer Disease , Animals , Mice , Humans , Alzheimer Disease/metabolism , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Neurons/metabolism , Plaque, Amyloid/metabolism , Disease Models, Animal
20.
Cells ; 10(8)2021 07 23.
Article in English | MEDLINE | ID: mdl-34440640

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder and results in severe neurodegeneration and progressive cognitive decline. Neurotrophins are growth factors involved in the development and survival of neurons, but also in underlying mechanisms for memory formation such as hippocampal long-term potentiation. Our aim was to identify small molecules with stimulatory effects on the signaling of two neurotrophins, the nerve growth factor (NGF) and the brain derived neurotrophic factor (BDNF). To identify molecules that could potentiate neurotrophin signaling, 25,000 molecules were screened, which led to the identification of the triazinetrione derivatives ACD855 (Ponazuril) and later on ACD856, as positive allosteric modulators of tropomyosin related kinase (Trk) receptors. ACD855 or ACD856 potentiated the cellular signaling of the neurotrophin receptors with EC50 values of 1.9 and 3.2 or 0.38 and 0.30 µM, respectively, for TrkA or TrkB. ACD855 increased acetylcholine levels in the hippocampus by 40% and facilitated long term potentiation in rat brain slices. The compounds acted as cognitive enhancers in a TrkB-dependent manner in several different behavioral models. Finally, the age-induced cognitive dysfunction in 18-month-old mice could be restored to the same level as found in 2-month-old mice after a single treatment of ACD856. We have identified a novel mechanism to modulate the activity of the Trk-receptors. The identification of the positive allosteric modulators of the Trk-receptors might have implications for the treatment of Alzheimer's diseases and other diseases characterized by cognitive impairment.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Nootropic Agents/pharmacology , Receptors, Nerve Growth Factor/agonists , Age Factors , Animals , Brain/enzymology , Brain/physiopathology , Cell Line, Tumor , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Humans , Male , Maze Learning/drug effects , Membrane Glycoproteins , Mice, Inbred C57BL , Motor Activity/drug effects , Protein-Tyrosine Kinases , Rats, Sprague-Dawley , Receptor, trkA/agonists , Receptor, trkA/metabolism , Receptor, trkB/agonists , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Small Molecule Libraries , Triazines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL