Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.538
2.
J Neurochem ; 168(4): e4, 2024 Apr.
Article En | MEDLINE | ID: mdl-38607972

Acetylcholine release from striatal cholinergic interneurons is controlled differently depending on the firing pattern (Published in JNC 167.1 issue) https://onlinelibrary.wiley.com/doi/10.1111/jnc.15950.


Acetylcholine , Corpus Striatum , Neostriatum , Interneurons , Cholinergic Agents/pharmacology
3.
Neurosci Lett ; 825: 137710, 2024 Mar 10.
Article En | MEDLINE | ID: mdl-38432355

Extensive experimental evidence points to neuroinflammation and oxidative stress as major pathogenic events that initiate and drive the neurodegenerative process. Monosodium glutamate (MSG) is a widely used food additive in processed foods known for its umami taste-enhancing properties. However, concerns about its potential adverse effects on the brain have been raised. Thus, the present study investigated the impact of MSG on lipopolysaccharide (LPS)-induced neurotoxicity in rat brains. Wistar rats weighing between 180 g and 200 g were randomly allocated into four groups: control (received distilled water), MSG (received 1.5 g/kg/day), LPS (received 250 µg/kg/day), and LPS + MSG (received LPS, 250 µg/kg, and MSG, 1.5 g/kg). LPS was administered intraperitoneally for 7 days while MSG was administered orally for 14 days. Our results showed that MSG exacerbated LPS-induced impairment in locomotor and exploratory activities in rats. Similarly, MSG exacerbated LPS-induced oxidative stress as evidenced by increased levels of malondialdehyde (MDA) with a concomitant decrease in levels of superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), and glutathione-s-transferase (GST) in the brain tissue. In addition, MSG potentiated LPS-induced neuroinflammation, as indicated by increased levels of pro-inflammatory cytokines such as interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) as well as myeloperoxidase (MPO) and nitric oxide (NO) in the brain. Moreover, MSG aggravated LPS-induced cholinergic dysfunction, as demonstrated by increased activity of acetylcholinesterase (AChE) in the brain. Further, we found a large number of degenerative neurons widespread in hippocampal CA1, CA3 regions, cerebellum, and cortex according to H&E staining. Taken together, our findings suggest that MSG aggravates LPS-induced neurobehavioral deficits, oxidative stress, neuroinflammation, cholinergic dysfunction, and neurodegeneration in rat brains.


Lipopolysaccharides , Sodium Glutamate , Rats , Animals , Sodium Glutamate/toxicity , Lipopolysaccharides/toxicity , Rats, Wistar , Acetylcholinesterase/metabolism , Neuroinflammatory Diseases , Oxidative Stress , Glutathione/metabolism , Brain/metabolism , Cholinergic Agents/pharmacology
4.
Pharmacol Biochem Behav ; 238: 173749, 2024 May.
Article En | MEDLINE | ID: mdl-38462045

BACKGROUND: Muscarinic or 5-HT1A receptors are crucial in learning and memory processes, and their expression is evident in the brain areas involved in cognition. The administration of the activators of these receptors prevents the development of cognitive dysfunctions in animal models of schizophrenia induced by MK-801 (N-methyl-d-aspartate receptor antagonist) administration. GABAergic dysfunction is considered as one of the most important causes of MK-801-induced spatial learning deficits. METHODS: Novel object recognition (NOR) and Morris water maze (MWM) tests were used to study the anti-amnestic effect of the biased 5-HT1A receptor agonist (F15599) alone or in combinations with VU0357017 (M1 receptor allosteric agonist), VU0152100 (M4 receptor positive allosteric modulator), and VU0238429 (M5 receptor positive allosteric modulator) on MK-801-induced dysfunctions. The compounds were administered for 5 consecutive days. Animals tested with the MWM underwent 5-day training. Western blotting was used to study the expressions of 5-HT1A receptors and the level of GAD65 in the frontal cortices (FCs) and hippocampi of the animals. RESULTS: F15599 prevented the amnestic effect induced by MK-801 in the MWM at a dose of 0.1 mg/kg. The co-administration of the compound with muscarinic receptors activators had no synergistic effect. The additive effect of the combinations was evident in the prevention of declarative memory dysfunctions investigated in NOR. The administration of MK-801 impaired 5-HT1A expression in the hippocampi and decreased GAD65 levels in both the FCs and hippocampi. The administration of muscarinic ligands prevented these MK-801-induced deficits only in the hippocampi of MWM-trained animals. No effects of the compounds were observed in untrained mice. CONCLUSION: Our results indicate that F15599 prevents schizophrenia-related spatial learning deficits in the MWM; however, the activity of the compound is not intensified with muscarinic receptors activators. In contrast, the combined administration of the ligands is effective in the NOR model of declarative memory. The muscarinic receptors activators reversed MK-801-induced 5-HT1A and GAD65 dysfunctions in the hippocampi of MWM-trained mice, but not in untrained mice.


Dizocilpine Maleate , Serotonin , Mice , Animals , Dizocilpine Maleate/pharmacology , Receptors, Muscarinic , Brain , Cholinergic Agents/pharmacology , Receptor, Serotonin, 5-HT1A
5.
Synapse ; 78(2): e22287, 2024 Mar.
Article En | MEDLINE | ID: mdl-38427384

Direct pathway striatal projection neurons (dSPNs) are characterized by the expression of dopamine (DA) class 1 receptors (D1 R), as well as cholinergic muscarinic M1 and M4 receptors (M1 R, M4 R). D1 R enhances neuronal firing through phosphorylation of voltage-gate calcium channels (CaV 1 Ca2+ channels) activating Gs proteins and protein kinase A (PKA). Concurrently, PKA suppresses phosphatase PP-1 through DARPP-32, thus extending this facilitatory modulation. M1 R also influences Ca2+ channels in SPNs through Gq proteins and protein kinase C. However, the signaling mechanisms of M4 R in dSPNs are less understood. Two pathways are attributed to M4 R: an inhibitory one through Gi/o proteins, and a facilitatory one via the cyclin Cdk5. Our study reveals that a previously observed facilitatory modulation via CaV 1 Ca2+ channels is linked to the Cdk5 pathway in dSPNs. This result could be significant in treating parkinsonism. Therefore, we questioned whether this effect persists post DA-depletion in experimental parkinsonism. Our findings indicate that in such conditions, M4 R activation leads to a decrease in Ca2+ current and an increased M4 R protein level, contrasting with the control response. Nevertheless, parkinsonian and control actions are inhibited by the Cdk5 inhibitor roscovitine, suggesting Cdk5's role in both conditions. Cdk5 may activate PP-1 via PKA inhibition in DA depletion. Indeed, we found that inhibiting PP-1 restores control M4 R actions, implying that PP-1 is overly active via M4 Rs in DA-depleted condition. These insights contribute to understanding how DA-depletion alters modulatory signaling in striatal neurons. Additional working hypotheses are discussed.


Corpus Striatum , Dopamine , Dopamine/metabolism , Corpus Striatum/metabolism , Interneurons/metabolism , Neurons/metabolism , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology
6.
Neurochem Res ; 49(5): 1137-1149, 2024 May.
Article En | MEDLINE | ID: mdl-38300457

Vascular dementia (VD) is a degenerative cerebrovascular disorder associated with progressive cognitive decline. Previous reports have shown that 7,8-dihydroxyflavone (7,8-DHF), a well-known TrkB agonist, effectively ameliorates cognitive deficits in several disease models. Therefore, this study investigated the protective effects of 7,8-DHF against 2-VO-induced VD. VD was established in rats using the permanent bilateral carotid arteries occlusion (two-vessel occlusion, 2-VO) model. 7,8-DHF (5, 10, and 20 mg/kg) and Donepezil (10 mg/kg) were administered for 4 weeks. Memory function was assessed by the novel objective recognition task (NOR) and Morris water maze (MWM) tests. Inflammatory (TNF-α, IL-1ß, and NF-kß), oxidative stress, and apoptotic (BAX, BCL-2, caspase-3) markers, along with the activity of choline acetylcholinesterase (AChE) was assessed. p-AKT, p-CREB, BDNF, and neurotransmitter (NT) (GLU, GABA, and ACh) levels were also analyzed in the hippocampus of 2-VO rats. Our results show that 7,8-DHF effectively improved memory performance and cholinergic dysfunction in 2-VO model rats. Furthermore, 7,8-DHF treatment also increased p-AKT, p-CREB, and BDNF levels, suppressed oxidative, inflammatory, and apoptotic markers, and restored altered NT levels in the hippocampus. These findings imply that 7, 8-DHF may act via multiple mechanisms and as such serve as a promising neuroprotective agent in the context of VD.


Dementia, Vascular , Rats , Animals , Dementia, Vascular/drug therapy , Acetylcholinesterase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Brain-Derived Neurotrophic Factor , Maze Learning , Oxidative Stress , Apoptosis , Inflammation/drug therapy , Hippocampus/metabolism , Cholinergic Agents/pharmacology
7.
J Neurosci ; 44(12)2024 Mar 20.
Article En | MEDLINE | ID: mdl-38320853

Gonadotropin-releasing hormone (GnRH)-synthesizing neurons orchestrate reproduction centrally. Early studies have proposed the contribution of acetylcholine (ACh) to hypothalamic control of reproduction, although the causal mechanisms have not been clarified. Here, we report that in vivo pharmacogenetic activation of the cholinergic system increased the secretion of luteinizing hormone (LH) in orchidectomized mice. 3DISCO immunocytochemistry and electron microscopy revealed the innervation of GnRH neurons by cholinergic axons. Retrograde viral labeling initiated from GnRH-Cre neurons identified the medial septum and the diagonal band of Broca as exclusive sites of origin for cholinergic afferents of GnRH neurons. In acute brain slices, ACh and carbachol evoked a biphasic effect on the firing rate in GnRH neurons, first increasing and then diminishing it. In the presence of tetrodotoxin, carbachol induced an inward current, followed by a decline in the frequency of miniature postsynaptic currents (mPSCs), indicating a direct influence on GnRH cells. RT-PCR and whole-cell patch-clamp studies revealed that GnRH neurons expressed both nicotinic (α4ß2, α3ß4, and α7) and muscarinic (M1-M5) AChRs. The nicotinic AChRs contributed to the nicotine-elicited inward current and the rise in firing rate. Muscarine via M1 and M3 receptors increased, while via M2 and M4 reduced the frequency of both mPSCs and firing. Optogenetic activation of channelrhodopsin-2-tagged cholinergic axons modified GnRH neuronal activity and evoked cotransmission of ACh and GABA from a subpopulation of boutons. These findings confirm that the central cholinergic system regulates GnRH neurons and activates the pituitary-gonadal axis via ACh and ACh/GABA neurotransmissions in male mice.


Acetylcholine , Gonadotropin-Releasing Hormone , Mice , Animals , Male , Acetylcholine/pharmacology , Carbachol/pharmacology , Neurons/physiology , Cholinergic Agents/pharmacology , Nicotine/pharmacology , Luteinizing Hormone , gamma-Aminobutyric Acid/pharmacology
8.
Eur J Pharmacol ; 968: 176430, 2024 Apr 05.
Article En | MEDLINE | ID: mdl-38369274

Memory impairment affects cognition and information processing, and attention, leading to a decline in life quality of patients. Previous studies have shown the memory-improving effects of sea cucumber peptides. This study further explored the memory-improving mechanisms of sea cucumber peptides using scopolamine-induced memory-impaired mice and identified novel memory-improving peptides within low molecular weight peptide fractions. The sea cucumber peptides were categorized into three groups based on their molecular weights: SCP-L (molecular weight greater than 10 kDa), SCP-M (weight between 3 kDa and 10 kDa), and SCP-S (molecular weight less than 3 kDa). The results showed that SCP-S improved behavioral performance by regulating cholinergic system disorder and reducing oxidative stress levels, distinguishing itself from SCP-M and SCP-L. Further, SCP-S was found to exhibit a well ability in alleviating the degree of neuroinflammation dependent on microglia and promoting synaptic plasticity. Additionally, a novel memory-improving peptide Ser-Phe-Gly-Asp-Ile (SFGDI) was identified by EASY-nano-LC/MS/MS after simulated digestion-absorption coupling of in silico technologies from SCP-S. SFGDI protected against oxidative stress and regulated cholinergic system in scopolamine-induced PC12 cells. These findings suggest that SCP-S and SFGDI might be considered as potential memory-improving food for people suffering from memory disorders.


Scopolamine , Sea Cucumbers , Rats , Humans , Mice , Animals , Scopolamine/pharmacology , Tandem Mass Spectrometry , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Peptides/pharmacology , Peptides/therapeutic use , Oxidative Stress , Cholinergic Agents/pharmacology
9.
Int J Mol Sci ; 25(3)2024 Feb 03.
Article En | MEDLINE | ID: mdl-38339131

Glucagon-like peptide-2 (GLP-2) has been reported to influence gastrointestinal motor responses, exerting a modulatory role on enteric neurotransmission. To our knowledge, no data on GLP-2 effects on the motility of the isolated ileum are available; therefore, we investigated whether GLP-2 affects the contractile activity of mouse ileal preparations and the neurotransmitters engaged. Ileal preparations showed tetrodotoxin (TTX)- and atropine-insensitive spontaneous contractile activity, which was unaffected by the nitric oxide synthesis inhibitor, L-NNA. GLP-2 depressed the spontaneous contractility, an effect that was abolished by TTX or L-NNA and not influenced by atropine. Electrical field stimulation induced TTX- and atropine-sensitive contractile responses, which were reduced in amplitude by GLP-2 even in the presence of L-NNA. Immunohistochemical results showed a significant increase in nNOS-positive fibers in the ileal muscle wall and a significant decrease in ChAT-positive myenteric neurons in GLP-2-exposed preparations. The present results offer the first evidence that GLP-2 acts on ileal preparations. The hormone appears to depress ileal contractility through a dual opposite modulatory effect on inhibitory nitrergic and excitatory cholinergic neurotransmission. From a physiological point of view, it could be hypothesized that GLP-2 inhibitory actions on ileal contractility can increase transit time, facilitating nutrient absorption.


Glucagon-Like Peptide 2 , Synaptic Transmission , Mice , Animals , Muscle Contraction/physiology , Nitroarginine/pharmacology , Ileum , Cholinergic Agents/pharmacology , Atropine Derivatives/pharmacology , Electric Stimulation
10.
ACS Chem Neurosci ; 15(5): 994-1009, 2024 Mar 06.
Article En | MEDLINE | ID: mdl-38407056

Cholinergic deficit is a characteristic factor of several pathologies, such as myasthenia gravis, some types of congenital myasthenic syndromes, and Alzheimer's Disease. Two molecular targets for its treatment are acetylcholinesterase (AChE) and nicotinic acetylcholine receptor (nAChR). In previous studies, we found that caffeine behaves as a partial nAChR agonist and confirmed that it inhibits AChE. Here, we present new bifunctional caffeine derivatives consisting of a theophylline ring connected to amino groups by different linkers. All of them were more potent AChE inhibitors than caffeine. Furthermore, although some of them also activated muscle nAChR as partial agonists, not all of them stabilized nAChR in its desensitized conformation. To understand the molecular mechanism underlying these results, we performed docking studies on AChE and nAChR. The nAChR agonist behavior of the compounds depends on their accessory group, whereas their ability to stabilize the receptor in a desensitized state depends on the interactions of the linker at the binding site. Our results show that the new compounds can inhibit AChE and activate nAChR with greater potency than caffeine and provide further information on the modulation mechanisms of pharmacological targets for the design of novel therapeutic interventions in cholinergic deficit.


Caffeine , Receptors, Nicotinic , Caffeine/pharmacology , Acetylcholinesterase/metabolism , Receptors, Nicotinic/metabolism , Cholinergic Agents/pharmacology , Cholinesterase Inhibitors/pharmacology
11.
J Neurosci ; 44(11)2024 Mar 13.
Article En | MEDLINE | ID: mdl-38286627

Dopamine neurons play crucial roles in pleasure, reward, memory, learning, and fine motor skills and their dysfunction is associated with various neuropsychiatric diseases. Dopamine receptors are the main target of treatment for neurologic and psychiatric disorders. Antipsychotics that antagonize the dopamine D2 receptor (DRD2) are used to alleviate the symptoms of these disorders but may also sometimes cause disabling side effects such as parkinsonism (catalepsy in rodents). Here we show that GPR143, a G-protein-coupled receptor for L-3,4-dihydroxyphenylalanine (L-DOPA), expressed in striatal cholinergic interneurons enhances the DRD2-mediated side effects of haloperidol, an antipsychotic agent. Haloperidol-induced catalepsy was attenuated in male Gpr143 gene-deficient (Gpr143-/y ) mice compared with wild-type (Wt) mice. Reducing the endogenous release of L-DOPA and preventing interactions between GPR143 and DRD2 suppressed the haloperidol-induced catalepsy in Wt mice but not Gpr143-/y mice. The phenotypic defect in Gpr143-/y mice was mimicked in cholinergic interneuron-specific Gpr143-/y (Chat-cre;Gpr143flox/y ) mice. Administration of haloperidol increased the phosphorylation of ribosomal protein S6 at Ser240/244 in the dorsolateral striatum of Wt mice but not Chat-cre;Gpr143flox/y mice. In Chinese hamster ovary cells stably expressing DRD2, co-expression of GPR143 increased cell surface expression level of DRD2, and L-DOPA application further enhanced the DRD2 surface expression. Shorter pauses in cholinergic interneuron firing activity were observed after intrastriatal stimulation in striatal slice preparations from Chat-cre;Gpr143flox/y mice compared with those from Wt mice. Together, these findings provide evidence that GPR143 regulates DRD2 function in cholinergic interneurons and may be involved in parkinsonism induced by antipsychotic drugs.


Antipsychotic Agents , Parkinsonian Disorders , Receptors, Neurotransmitter , Humans , Mice , Male , Animals , Cricetinae , Haloperidol/pharmacology , Levodopa/adverse effects , Catalepsy/chemically induced , CHO Cells , Cricetulus , Antipsychotic Agents/adverse effects , Interneurons/metabolism , Cholinergic Agents/pharmacology , Eye Proteins/metabolism , Membrane Glycoproteins/metabolism
12.
Eur J Pharmacol ; 965: 176332, 2024 Feb 15.
Article En | MEDLINE | ID: mdl-38228217

Alzheimer's disease (AD) is the most common form of dementia and is a progressive neurodegenerative disorder of the brain. Most AD experimental animal models are pharmacological or transgenic in origin. The existing pharmacological approaches for developing AD are poorly developed and most of them fail to replicate the complete characteristics of disease pathology. Developing a cost-effective and reliable experimental animal model will meet this research gap. Zebrafish (ZF) are progressively emerging as a powerful drug discovery disease model to evaluate central nervous system (CNS) disorders due to their homologous similarities to humans as well as cost-effectiveness. The present research is conceptualized to develop and evaluate a reliable ZF AD model using aluminum chloride (AlCl3). Chronic exposure of 0.04 mM of AlCl3 for 28 days increased the expression of amyloid-ß, phosphorylated tau protein and senile plaque development in the ZF brain. The observed changes were associated with learning and memory impairment. Furthermore, decreased brain-derived neurotrophic factor (BDNF) level and elevated oxidative stress indices, pro-inflammatory cytokines levels and acetylcholine esterase (AChE) activity was observed upon exposure to AlCl3 in the ZF brain. Chronic exposure to 0.04 mM of AlCl3 would be a cost-effective ZF AD model for pharmacological screening and may also be used to unravel the molecular mechanism underlying the neuropathology of the disease.


Alzheimer Disease , Humans , Animals , Aluminum Chloride , Alzheimer Disease/metabolism , Zebrafish , Chlorides/toxicity , Neuroinflammatory Diseases , Oxidative Stress , Cholinergic Agents/pharmacology , Disease Models, Animal
13.
Biomed Pharmacother ; 171: 116190, 2024 Feb.
Article En | MEDLINE | ID: mdl-38278026

Sinomenine (SIN), an alkaloid extracted from the Chinese herbal medicine Sinomenium acutum, has great potential in anti-inflammatory, immune regulation, analgesic and sedative, and is already a clinical drug for the treatment of rheumatoid arthritis in China. Our previous studies show SIN inhibits inflammation by regulating ɑ7nAChR, a key receptor of cholinergic anti-inflammatory pathway (CAP), which plays an important role in regulating peripheral and central nervous system inflammation. Growing evidence supports the cholinergic dysregulation and inflammatory responses play the key role in the pathogenesis of AD. The intervention effects of SIN on AD by regulating CAP and homeostasis in brain and gut were analyzed for the first time in the present study using scopolamine-induced AD model mice. Behavioral tests were used to assess the cognitive performance. The neurons loss, cholinergic function, inflammation responses, biological barrier function in the mouse brain and intestinal tissues were evaluated through a variety of techniques, and the gut microbiota was detected using 16SrRNA sequencing. The results showed that SIN significantly inhibited the cognitive decline, dysregulation of cholinergic system, peripheral and central inflammation, biological barrier damage as well as intestinal flora disturbance caused by SCOP in mice. More importantly, SIN effectively regulated CAP to suppress the activation of TLR4/NF-κB and protect the homeostasis in brain and gut to alleviate cognitive impairment.


Alzheimer Disease , Morphinans , NF-kappa B , Mice , Animals , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Neuroimmunomodulation , Scopolamine/pharmacology , Inflammation/pathology , Homeostasis , Brain/metabolism , Cholinergic Agents/pharmacology
14.
Toxicon ; 239: 107595, 2024 Feb 23.
Article En | MEDLINE | ID: mdl-38211804

The neurotoxic impact of dietary exposure to aflatoxin B1 (AFB1) is well documented in experimental studies. Rutin is a phytochemical with prominent anti-inflammatory and antioxidant activities. There is an information gap on the influence of rutin on AFB1-induced neurotoxicity. This study investigated the influence of rutin on neurobehavioral and biochemical abnormalities in male Wistar rats (six weeks old) orally treated with AFB1 (0.75, and 1.5 mg/kg body weight) or co-administered with rutin (50 mg/kg) for 30 uninterrupted days. Results indicate that AFB1-induced depression-like behavior by Tail Suspension Test (TST) and cognitive impairment by Y-maze was abated following rutin co-administration. Abatement of AFB1-induced decreases in acetylcholinesterase (AChE) activity, and increased antioxidant status, by rutin was accompanied by a marked reduction in oxidative stress markers and increased hydrolysis of the purinergic molecules in the cerebral cortex and hippocampus of rats. Additionally, rutin co-treatment abrogated AFB1-mediated elevation of interleukin-6 (IL-6), nitric oxide (NO) levels, and activity of myeloperoxidase (MPO). Correspondingly, rutin co-treatment lowered the activity and immunocontent of immunosuppressive indoleamine 2, 3-dioxygenase (IDO). Further, rutin co-treatment prevented histological injuries in the cerebral cortex and hippocampus. In conclusion, abatement of AFB1-induced neurobehavioral abnormalities by rutin involves the mechanisms of anti-inflammatory, antioxidant, and regulation of cholinergic, purinergic, and indoleaminergic pathways in rats.


Aflatoxin B1 , Antioxidants , Rats , Male , Animals , Rats, Wistar , Aflatoxin B1/toxicity , Antioxidants/pharmacology , Antioxidants/metabolism , Rutin/pharmacology , Acetylcholinesterase , Hippocampus , Cerebral Cortex/metabolism , Oxidative Stress , Oxidation-Reduction , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology , Anti-Inflammatory Agents/pharmacology
15.
Environ Pollut ; 344: 123327, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38190878

The definition of microplastics (MPs) is nowadays too generic from a biological perspective, since different characteristics of these particles might influence their effects. To provide experimental evidence that size is an important factor to be considered, Mediterranean mussels Mytilus galloprovincialis were exposed to five size classes of polyethylene fragments (PE-MPs, 20-50 µm, 50-100 µm, 100-250 µm, 250-500 µm, 500-1000 µm). After 10 days of exposure, MPs ingestion and mechanistic relationships between particles size and cellular effects were analysed through a wide panel of biological alterations, including immune system responses, cholinergic function, antioxidant system, lipid metabolism and peroxidation. Results were further elaborated through a Weight of Evidence approach, summarizing the overall biological significance of obtained results in a hazard index based on the number and magnitude of variations and their toxicological relevance. PE-MPs 500-1000 µm were identified as the less biologically reactive size class due to the limited ingestion of particles coupled with the lack of biological effects, followed by PE-MPs 250-500 µm, which slightly altered the cholinergic function and lysosomal membranes. Conversely, PE-MPs smaller than 250 µm provoked a more consistent onset of biological alterations in terms of immune system composition and functioning, redox homeostasis, and lipid metabolism. The overall findings of this study highlight the importance of considering the size of particles for monitoring and risk assessment of MPs, introducing a more integrated evaluation of plastic pollution that, beside particles concentration, should adequately weigh those characteristics triggering the onset of biological effects.


Mytilus , Water Pollutants, Chemical , Animals , Microplastics/analysis , Plastics/analysis , Mytilus/metabolism , Polyethylene/metabolism , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology , Water Pollutants, Chemical/analysis
16.
Development ; 151(1)2024 Jan 01.
Article En | MEDLINE | ID: mdl-38063486

Cholinergic signaling plays a crucial role in the regulation of adult hippocampal neurogenesis; however, the mechanisms by which acetylcholine mediates neurogenic effects are not completely understood. Here, we report the expression of muscarinic acetylcholine receptor subtype M4 (M4 mAChR) on a subpopulation of neural precursor cells (NPCs) in the adult mouse hippocampus, and demonstrate that its pharmacological stimulation promotes their proliferation, thereby enhancing the production of new neurons in vivo. Using a targeted ablation approach, we also show that medial septum (MS) and the diagonal band of Broca (DBB) cholinergic neurons support both the survival and morphological maturation of adult-born neurons in the mouse hippocampus. Although the systemic administration of an M4-selective allosteric potentiator fails to fully rescue the MS/DBB cholinergic lesion-induced decrease in hippocampal neurogenesis, it further exacerbates the impairment in the morphological maturation of adult-born neurons. Collectively, these findings reveal stage-specific roles of M4 mAChRs in regulating adult hippocampal neurogenesis, uncoupling their positive role in enhancing the production of new neurons from the M4-induced inhibition of their morphological maturation, at least in the context of cholinergic signaling dysfunction.


Neural Stem Cells , Receptor, Muscarinic M4 , Mice , Animals , Receptor, Muscarinic M4/metabolism , Neural Stem Cells/metabolism , Hippocampus/metabolism , Neurogenesis/genetics , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology , Cell Proliferation
17.
J Neurochem ; 168(4): 334-338, 2024 Apr.
Article En | MEDLINE | ID: mdl-38082541

This preface introduces the Journal of Neurochemistry special issue on Cholinergic Mechanisms that highlights the progress in the molecular, structural, neurochemical, pharmacological, toxicological, and clinical studies of the cholinergic system which underline its complexity and impact on health and disease. This issue comprises of (systematic) reviews and original articles, the majority of which have been presented at the 17th International Symposium on Cholinergic Mechanisms (ISCM2022) held in Dubrovnik, Croatia in May 2022. The symposium brought together leading "Cholinergikers" to shed new light on cholinergic transmission, ranging from the molecular to the clinical and cognitive mechanisms.


Acetylcholine , Receptors, Nicotinic , Acetylcholine/pharmacology , Synaptic Transmission , Cholinergic Agents/pharmacology , Receptors, Nicotinic/metabolism
18.
Brain Res ; 1823: 148704, 2024 01 15.
Article En | MEDLINE | ID: mdl-38052316

Alzheimer's disease (AD) is a chronic, progressive neurodegenerative condition marked by cognitive impairment. Although coconut oil has been shown to be potentially beneficial in reducing AD-related cognitive deficits, information on its mechanism of action is limited. Thus, we investigated the effects of coconut oil on spatial cognitive ability and non-cognitive functions in a rat model of AD induced by G-galactose (D-GAL) and aluminum chloride (AlCl3), and examined the changes in synaptic transmission, cholinergic activity, neurotrophic factors and oxidative stress in this process. The AD model was established by administering D-GAL and AlCl3 for 90 days, while also supplementing with coconut oil during this time. Cognitive and non-cognitive abilities of the rats were evaluated at the end of the 90-day supplementation period. In addition, biochemical markers related to the pathogenesis of the AD were measures in the hippocampus tissue. Exposure to D-GAL/AlCl3 resulted in a reduction in locomotor activity, an elevation in anxiety-like behavior, and an impairment of spatial learning and memory (P < 0.05). The aforementioned behavioral disturbances were observed to coincide with increased oxidative stress and cholinergic impairment, as well as reduced synaptic transmission and levels of neurotrophins in the hippocampus (P < 0.05). Interestingly, treatment with coconut oil attenuated all the neuropathological changes mentioned above (P < 0.05). These findings suggest that coconut oil shows protective effects against cognitive and non-cognitive impairment, AD pathology markers, oxidative stress, synaptic transmission, and cholinergic function in a D-GAL/AlCl3-induced AD rat model.


Alzheimer Disease , Cognition Disorders , Cognitive Dysfunction , Neuroprotective Agents , Rats , Animals , Coconut Oil/pharmacology , Aluminum Chloride/adverse effects , Cognition Disorders/drug therapy , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/pathology , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Hippocampus , Oxidative Stress , Cholinergic Agents/pharmacology , Disease Models, Animal , Galactose/toxicity , Neuroprotective Agents/therapeutic use
19.
Poult Sci ; 103(2): 103275, 2024 Feb.
Article En | MEDLINE | ID: mdl-38042041

Atrial and ventricular myocardium from young (6-wk-old), young adult (3-6-mo-old), and aged (14-15-mo-old) meat-type (B.U.T. Big 6) and wild-type (Cröllwitzer) turkeys were used to study the influence of age and sex on cholinergic muscarinic receptors using [3H]-N-methyl-scopolamine (3H-NMS) binding studies. In both breeds, saturation experiments indicated the presence of regional-, sex-, and age-related differences in the density of cholinergic muscarinic receptors (Bmax), that is, a decrease or increase. Except for right atria, Bmax was decreased in both male and female B.U.T. Big 6 hearts with increasing age. Similarly, a negative correlation between Bmax and age could be seen in female and male atria of Cröllwitzer turkeys, while positive correlation could be seen in right and left ventricles of male, and only right ventricles of female Cröllwitzer turkeys. The affinity of the receptor (KD) was not affected by age, sex and breed. In all cardiac chamber tissues, the M2-subtype was shown to be predominant followed by the M3-subtype and to a lesser extent the M1-subtype. Aspects of this age-dependent remodeling of the heart differ between sexes, resulting in maladaptive changes in older turkeys with a high degree of frailty. These observations may help explain why males and females are susceptible to different cardiovascular diseases as they age and why frail older adults are most often affected by these diseases.


Heart Ventricles , Turkeys , Male , Female , Animals , Turkeys/metabolism , Chickens/metabolism , Heart Atria , Receptors, Muscarinic/metabolism , Myocardium/metabolism , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology
20.
J Neurosci ; 44(3)2024 Jan 17.
Article En | MEDLINE | ID: mdl-38050146

Acetylcholine (ACh) promotes neocortical output to the thalamus and brainstem by preferentially enhancing the postsynaptic excitability of layer 5 pyramidal tract (PT) neurons relative to neighboring intratelencephalic (IT) neurons. Less is known about how ACh regulates the excitatory synaptic drive of IT and PT neurons. To address this question, spontaneous excitatory postsynaptic potentials (sEPSPs) were recorded in dual recordings of IT and PT neurons in slices of prelimbic cortex from adult female and male mice. ACh (20 µM) enhanced sEPSP amplitudes, frequencies, rise-times, and half-widths preferentially in PT neurons. These effects were blocked by the muscarinic receptor antagonist atropine (1 µM). When challenged with pirenzepine (1 µM), an antagonist selective for M1-type muscarinic receptors, ACh instead reduced sEPSP frequencies, suggesting that ACh may generally suppress synaptic transmission in the cortex via non-M1 receptors. Cholinergic enhancement of sEPSPs in PT neurons was not sensitive to antagonism of GABA receptors with gabazine (10 µM) and CGP52432 (2.5 µM) but was blocked by tetrodotoxin (1 µM), suggesting that ACh enhances action-potential-dependent excitatory synaptic transmission in PT neurons. ACh also preferentially promoted the occurrence of synchronous sEPSPs in dual recordings of PT neurons relative to IT-PT and IT-IT parings. Finally, selective chemogenetic silencing of hM4Di-expressing PT, but not commissural IT, neurons blocked cholinergic enhancement of sEPSP amplitudes and frequencies in PT neurons. These data suggest that, in addition to selectively enhancing the postsynaptic excitability of PT neurons, M1 receptor activation promotes corticofugal output by amplifying recurrent excitation within networks of PT neurons.


Cholinergic Agents , Neurons , Mice , Male , Female , Animals , Cholinergic Agents/pharmacology , Neurons/physiology , Pyramidal Cells/physiology , Synaptic Transmission/physiology , Acetylcholine/pharmacology , Prefrontal Cortex/physiology , Receptor, Muscarinic M1
...