Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.620
Filter
2.
Hear Res ; 447: 109025, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733712

ABSTRACT

Cortical acetylcholine (ACh) release has been linked to various cognitive functions, including perceptual learning. We have previously shown that cortical cholinergic innervation is necessary for accurate sound localization in ferrets, as well as for their ability to adapt with training to altered spatial cues. To explore whether these behavioral deficits are associated with changes in the response properties of cortical neurons, we recorded neural activity in the primary auditory cortex (A1) of anesthetized ferrets in which cholinergic inputs had been reduced by making bilateral injections of the immunotoxin ME20.4-SAP in the nucleus basalis (NB) prior to training the animals. The pattern of spontaneous activity of A1 units recorded in the ferrets with cholinergic lesions (NB ACh-) was similar to that in controls, although the proportion of burst-type units was significantly lower. Depletion of ACh also resulted in more synchronous activity in A1. No changes in thresholds, frequency tuning or in the distribution of characteristic frequencies were found in these animals. When tested with normal acoustic inputs, the spatial sensitivity of A1 neurons in the NB ACh- ferrets and the distribution of their preferred interaural level differences also closely resembled those found in control animals, indicating that these properties had not been altered by sound localization training with one ear occluded. Simulating the animals' previous experience with a virtual earplug in one ear reduced the contralateral preference of A1 units in both groups, but caused azimuth sensitivity to change in slightly different ways, which may reflect the modest adaptation observed in the NB ACh- group. These results show that while ACh is required for behavioral adaptation to altered spatial cues, it is not required for maintenance of the spectral and spatial response properties of A1 neurons.


Subject(s)
Acoustic Stimulation , Auditory Cortex , Basal Forebrain , Ferrets , Animals , Auditory Cortex/metabolism , Auditory Cortex/physiopathology , Basal Forebrain/metabolism , Sound Localization , Acetylcholine/metabolism , Male , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Auditory Pathways/physiopathology , Auditory Pathways/metabolism , Female , Immunotoxins/toxicity , Basal Nucleus of Meynert/metabolism , Basal Nucleus of Meynert/physiopathology , Basal Nucleus of Meynert/pathology , Neurons/metabolism , Auditory Threshold , Adaptation, Physiological , Behavior, Animal
3.
Elife ; 132024 May 15.
Article in English | MEDLINE | ID: mdl-38748470

ABSTRACT

Acetylcholine is widely believed to modulate the release of dopamine in the striatum of mammals. Experiments in brain slices clearly show that synchronous activation of striatal cholinergic interneurons is sufficient to drive dopamine release via axo-axonal stimulation of nicotinic acetylcholine receptors. However, evidence for this mechanism in vivo has been less forthcoming. Mohebi, Collins and Berke recently reported that, in awake behaving rats, optogenetic activation of striatal cholinergic interneurons with blue light readily evokes dopamine release measured with the red fluorescent sensor RdLight1 (Mohebi et al., 2023). Here, we show that blue light alone alters the fluorescent properties of RdLight1 in a manner that may be misconstrued as phasic dopamine release, and that this artefactual photoactivation can account for the effects attributed to cholinergic interneurons. Our findings indicate that measurements of dopamine using the red-shifted fluorescent sensor RdLight1 should be interpreted with caution when combined with optogenetics. In light of this and other publications that did not observe large acetylcholine-evoked dopamine transients in vivo, the conditions under which such release occurs in behaving animals remain unknown.


Subject(s)
Cholinergic Neurons , Dopamine , Interneurons , Optogenetics , Dopamine/metabolism , Animals , Interneurons/metabolism , Interneurons/physiology , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Rats , Optogenetics/methods , Motivation , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Acetylcholine/metabolism
4.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38755010

ABSTRACT

Cholinergic neurons of the basal forebrain represent the main source of cholinergic innervation of large parts of the neocortex and are involved in adults in the modulation of attention, memory, and arousal. During the first postnatal days, they play a crucial role in the development of cortical neurons and cortical cytoarchitecture. However, their characteristics, during this period have not been studied. To understand how they can fulfill this role, we investigated the morphological and electrophysiological maturation of cholinergic neurons of the substantia innominata-nucleus basalis of Meynert (SI/NBM) complex in the perinatal period in mice. We show that cholinergic neurons, whether or not they express gamma-aminobutyric acid (GABA) as a cotransmitter, are already functional at Embryonic Day 18. Until the end of the first postnatal week, they constitute a single population of neurons with a well developed dendritic tree, a spontaneous activity including bursting periods, and a short-latency response to depolarizations (early-firing). They are excited by both their GABAergic and glutamatergic afferents. During the second postnatal week, a second, less excitable, neuronal population emerges, with a longer delay response to depolarizations (late-firing), together with the hyperpolarizing action of GABAA receptor-mediated currents. This classification into early-firing (40%) and late-firing (60%) neurons is again independent of the coexpression of GABAergic markers. These results strongly suggest that during the first postnatal week, the specific properties of developing SI/NBM cholinergic neurons allow them to spontaneously release acetylcholine (ACh), or ACh and GABA, into the developing cortex.


Subject(s)
Basal Forebrain , Cholinergic Neurons , gamma-Aminobutyric Acid , Animals , Cholinergic Neurons/physiology , Cholinergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Basal Forebrain/physiology , Basal Forebrain/metabolism , Animals, Newborn , Mice, Inbred C57BL , Female , Basal Nucleus of Meynert/physiology , Basal Nucleus of Meynert/metabolism , Substantia Innominata/physiology , Substantia Innominata/metabolism , Mice , Receptors, GABA-A/metabolism , Action Potentials/physiology , Patch-Clamp Techniques , Glutamic Acid/metabolism
5.
J Alzheimers Dis ; 99(2): 639-656, 2024.
Article in English | MEDLINE | ID: mdl-38728184

ABSTRACT

Background: Familial Alzheimer's disease (FAD) presenilin 1 E280A (PSEN 1 E280A) is characterized by functional impairment and the death of cholinergic neurons as a consequence of amyloid-ß (Aß) accumulation and abnormal phosphorylation of the tau protein. Currently, there are no available therapies that can cure FAD. Therefore, new therapies are urgently needed for treating this disease. Objective: To assess the effect of sildenafil (SIL) on cholinergic-like neurons (ChLNs) harboring the PSEN 1 E280A mutation. Methods: Wild-type (WT) and PSEN 1 E280A ChLNs were cultured in the presence of SIL (25µM) for 24 h. Afterward, proteinopathy, cell signaling, and apoptosis markers were evaluated via flow cytometry and fluorescence microscopy. Results: We found that SIL was innocuous toward WT PSEN 1 ChLNs but reduced the accumulation of intracellular Aß fragments by 87%, decreased the non-physiological phosphorylation of the protein tau at residue Ser202/Thr205 by 35%, reduced the phosphorylation of the proapoptotic transcription factor c-JUN at residue Ser63/Ser73 by 63%, decreased oxidized DJ-1 at Cys106-SO3 by 32%, and downregulated transcription factor TP53 (tumor protein p53), BH-3-only protein PUMA (p53 upregulated modulator of apoptosis), and cleaved caspase 3 (CC3) expression by 20%, 32%, and 22%, respectively, compared with untreated mutant ChLNs. Interestingly, SIL also ameliorated the dysregulation of acetylcholine-induced calcium ion (Ca2+) influx in PSEN 1 E280A ChLNs. Conclusions: Although SIL showed no antioxidant capacity in the oxygen radical absorbance capacity and ferric ion reducing antioxidant power assays, it might function as an anti-amyloid and antiapoptotic agent and functional neuronal enhancer in PSEN 1 E280A ChLNs. Therefore, the SIL has therapeutic potential for treating FAD.


Subject(s)
Alzheimer Disease , Cholinergic Neurons , Mutation , Presenilin-1 , Sildenafil Citrate , Presenilin-1/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Mutation/genetics , Animals , Sildenafil Citrate/pharmacology , Amyloid beta-Peptides/metabolism , Humans , Cells, Cultured , Mice , tau Proteins/metabolism , tau Proteins/genetics , Phosphorylation/drug effects , Phenotype
6.
Cell Rep ; 43(5): 114187, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38722743

ABSTRACT

The locomotor role of dopaminergic neurons is traditionally attributed to their ascending projections to the basal ganglia, which project to the mesencephalic locomotor region (MLR). In addition, descending dopaminergic projections to the MLR are present from basal vertebrates to mammals. However, the neurons targeted in the MLR and their behavioral role are unknown in mammals. Here, we identify genetically defined MLR cells that express D1 or D2 receptors and control different motor behaviors in mice. In the cuneiform nucleus, D1-expressing neurons promote locomotion, while D2-expressing neurons stop locomotion. In the pedunculopontine nucleus, D1-expressing neurons promote locomotion, while D2-expressing neurons evoke ipsilateral turns. Using RNAscope, we show that MLR dopamine-sensitive neurons comprise a combination of glutamatergic, GABAergic, and cholinergic neurons, suggesting that different neurotransmitter-based cell types work together to control distinct behavioral modules. Altogether, our study uncovers behaviorally relevant cell types in the mammalian MLR based on the expression of dopaminergic receptors.


Subject(s)
Dopamine , Dopaminergic Neurons , Locomotion , Mesencephalon , Receptors, Dopamine D1 , Animals , Mesencephalon/metabolism , Mice , Dopaminergic Neurons/metabolism , Dopamine/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Mice, Inbred C57BL , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , GABAergic Neurons/metabolism , Male
7.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732223

ABSTRACT

Alzheimer's disease (AD) is characterized by a loss of neurons in the cortex and subcortical regions. Previously, we showed that the progressive degeneration of subcortical monoaminergic (MAergic) neurons seen in human AD is recapitulated in the APPswe/PS1ΔE9 (APP/PS) transgenic mouse model. Because degeneration of cholinergic (Ach) neurons is also a prominent feature of AD, we examined the integrity of the Ach system in the APP/PS model. The overall density of Ach fibers is reduced in APP/PS1 mice at 12 and 18 months of age but not at 4 months of age. Analysis of basal forebrain Ach neurons shows no loss of Ach neurons in the APP/PS model. Thus, since MAergic systems show overt cell loss at 18 months of age, the Ach system is less vulnerable to neurodegeneration in the APP/PS1 model. We also examined whether the proximity to Aß deposition affected the degeneration of Ach and 5-HT afferents. We found that the areas closer to the edges of compact Aß deposits exhibit a more severe loss of afferents than the areas that are more distal to Aß deposits. Collectively, the results indicate that the APP/PS model recapitulates the degeneration of multiple subcortical neurotransmitter systems, including the Ach system. In addition, the results indicate that Aß deposits cause global as well as local toxicity to subcortical afferents.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Cholinergic Neurons , Disease Models, Animal , Plaque, Amyloid , Presenilin-1 , Animals , Humans , Mice , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Mice, Transgenic , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism
8.
Cell Rep ; 43(5): 114204, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38748878

ABSTRACT

Amyotrophic lateral sclerosis can be caused by abnormal accumulation of TAR DNA-binding protein 43 (TDP-43) in the cytoplasm of neurons. Here, we use a C. elegans model for TDP-43-induced toxicity to identify the biological mechanisms that lead to disease-related phenotypes. By applying deep behavioral phenotyping and subsequent dissection of the neuromuscular circuit, we show that TDP-43 worms have profound defects in GABA neurons. Moreover, acetylcholine neurons appear functionally silenced. Enhancing functional output of repressed acetylcholine neurons at the level of, among others, G-protein-coupled receptors restores neurotransmission, but inefficiently rescues locomotion. Rebalancing the excitatory-to-inhibitory ratio in the neuromuscular system by simultaneous stimulation of the affected GABA- and acetylcholine neurons, however, not only synergizes the effects of boosting individual neurotransmitter systems, but instantaneously improves movement. Our results suggest that interventions accounting for the altered connectome may be more efficient in restoring motor function than those solely focusing on diseased neuron populations.


Subject(s)
Caenorhabditis elegans , DNA-Binding Proteins , Disease Models, Animal , Animals , Caenorhabditis elegans/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , GABAergic Neurons/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Motor Neurons/metabolism , Locomotion , Synaptic Transmission , Movement , Cholinergic Neurons/metabolism
9.
Cells ; 13(7)2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38607082

ABSTRACT

Basal forebrain cholinergic dysfunction, most likely linked with tau protein aggregation, is a characteristic feature of Alzheimer's disease (AD). Recent evidence suggests that tau protein is a putative target for the treatment of dementia, and the tau aggregation inhibitor, hydromethylthionine mesylate (HMTM), has emerged as a potential disease-modifying treatment. However, its efficacy was diminished in patients already receiving approved acetylcholinesterase inhibitors. In this study, we ask whether this negative interaction can also be mimicked in experimental tau models of AD and whether the underlying mechanism can be understood. From a previous age profiling study, 6-month-old line 1 (L1) tau transgenic mice were characterized by a severe reduction in several cholinergic markers. We therefore assessed whether long-term pre-exposure with the acetylcholinesterase inhibitor rivastigmine alone and in conjunction with the tau aggregation inhibitor HMTM can reverse cholinergic deficits in L1. Rivastigmine and HMTM, and combinations of the two compounds were administered orally for 11 weeks to both L1 and wild-type mice. The brains were sectioned with a focus on the basal forebrain, motor cortex and hippocampus. Immunohistochemical staining and quantification of choline acetyltransferase (ChAT), tyrosine kinase A (TrkA)-positive neurons and relative optical intensity (ROI) for vesicular acetylcholine transporter (VAChT), and acetylcholinesterase (AChE) reactivity confirmed reversal of the diminished cholinergic phenotype of interneurons (nucleus accumbens, striatum) and projection neurons (medial septum, nucleus basalis magnocellularis) by HMTM, to a greater extent than by rivastigmine alone in L1 mice. Combined administration did not yield additivity but, in most proxies, led to antagonistic effects in which rivastigmine decreased the benefits shown with HMTM alone. Local markers (VAChT and AChE) in target structures of the basal forebrain, motor cortex and hippocampal CA3 seemed to be normalized by HMTM, but not by rivastigmine or the combination of both drugs. HMTM, which was developed as a tau aggregation inhibitor, strongly decreased the tau load in L1 mice, however, not in combination with rivastigmine. Taken together, these data confirm a cholinergic phenotype in L1 tau transgenic mice that resembles the deficits observed in AD patients. This phenotype is reversible by HMTM, but at the same time appears to be subject to a homeostatic regulation induced by chronic pre-treatment with an acetylcholinesterase inhibitor, which interferes with the efficacy of HMTM. The strongest phenotypic reversal coincided with a normalization of the tau load in the cortex and hippocampus of L1, suggesting that tau accumulation underpins the loss of cholinergic markers in the basal forebrain and its projection targets.


Subject(s)
Alzheimer Disease , Tauopathies , Humans , Mice , Animals , Infant , Rivastigmine/pharmacology , Alzheimer Disease/metabolism , tau Proteins/metabolism , Cholinesterase Inhibitors/pharmacology , Acetylcholinesterase/metabolism , Neuroprotection , Cholinergic Neurons/metabolism , Tauopathies/drug therapy , Cholinergic Agents , Mice, Transgenic
10.
J Neurochem ; 168(4): e5, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38607973

ABSTRACT

Whole-brain neural connectivity to cholinergic neurons in the nucleus basalis of Meynert (Published in JNC 166.2 issue) https://onlinelibrary.wiley.com/doi/10.1111/jnc.15873.


Subject(s)
Basal Nucleus of Meynert , Cholinergic Neurons , Brain
12.
Eur J Neurosci ; 59(11): 3061-3073, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38576223

ABSTRACT

The present study aimed to examine the effect of cholinergic interneuron lesions in the dorsal striatum on duration-memory formation. Cholinergic interneurons in the dorsal striatum may be involved in the formation of duration memory since they are among the main inputs to the dorsal striatal muscarinic acetylcholine-1 receptors, which play a role in the consolidation of duration memory. Rats were sufficiently trained using a peak-interval 20 s procedure and then infused with anti-choline acetyltransferase-saporin into the dorsal striatum to cause selective ablation of cholinergic interneurons. To make the rats acquire new duration-memories, we trained them with a peak interval 40 s after lesion. Before lesion, the peak times (an index of duration memory) for sham-lesioned and lesioned groups were similar at approximately 20 s. In the peak interval 40 s session, the peak times for the sham-lesioned and lesioned groups were approximately 30 and 20 s, respectively. After additional peak interval 40 s sessions, the peak times of both groups were shifted to approximately 40 s. Those results suggest that the cholinergic interneuron lesion delayed new duration-memory acquisition. Subsequent experiments showed that cholinergic interneuron lesions did not retard the shift of peak time to the original target time (20 s). Following experiment without changing the target time after lesion showed that cholinergic interneuron lesions did not change their peak times. Our findings suggest that cholinergic interneurons in the dorsal striatum are involved in new duration-memory acquisition but not in the utilization of already acquired duration memory and interval timing.


Subject(s)
Cholinergic Neurons , Corpus Striatum , Interneurons , Animals , Interneurons/physiology , Male , Rats , Corpus Striatum/physiology , Cholinergic Neurons/physiology , Cholinergic Neurons/metabolism , Memory/physiology , Choline O-Acetyltransferase/metabolism , Rats, Wistar
13.
Biochem Pharmacol ; 224: 116201, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608783

ABSTRACT

Intestinal barrier dysfunction, leaky gut, is implicated in various diseases, including irritable bowel syndrome (IBS) and neurodegenerative conditions like Alzheimer's disease. Our recent investigation revealed that basal forebrain cholinergic neurons (BFCNs), critical for cognitive function, receive signals from butyrate and orexin, playing a role in regulating intestinal barrier function through adenosine A2B signaling and the vagus. This study explores the involvement and function of brain histamine, linked to BFCNs, in the regulation of intestinal barrier function. Colonic permeability, assessed by quantifying absorbed Evans blue in rat colonic tissue, showed that histamine did not affect increased colonic permeability induced by LPS when administered subcutaneously. However, intracisternal histamine administration improved colonic hyperpermeability. Elevating endogenous histamine levels in the brain with SKF91488, a histamine N-methyltransferase inhibitor, also improved colonic hyperpermeability. This effect was abolished by intracisternal chlorpheniramine, an histamine H1 receptor antagonist, not ranitidine, an H2 receptor antagonist. The SKF91488-induced improvement in colonic hyperpermeability was blocked by vagotomy, intracisternal pirenzepine (suppressing BFCNs activity), or alloxazine (an adenosine A2B receptor antagonist). Additionally, intracisternal chlorpheniramine injection eliminated butyrate-induced improvement in colonic hyperpermeability. These findings suggest that brain histamine, acting via the histamine H1 receptor, regulates intestinal barrier function involving BFCNs, adenosine A2B signaling, and the vagus. Brain histamine appears to centrally regulate intestinal barrier function influenced by butyrate, differentiating its actions from peripheral histamine in conditions like IBS, where mast cell-derived histamine induces leaky gut. Brain histamine emerges as a potential pharmacological target for diseases associated with leaky gut, such as dementia and IBS.


Subject(s)
Cholinergic Neurons , Colon , Histamine , Permeability , Rats, Sprague-Dawley , Receptor, Adenosine A2B , Vagus Nerve , Animals , Histamine/metabolism , Histamine/pharmacology , Rats , Male , Receptor, Adenosine A2B/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Vagus Nerve/drug effects , Vagus Nerve/physiology , Vagus Nerve/metabolism , Colon/metabolism , Colon/drug effects , Permeability/drug effects , Prosencephalon/drug effects , Prosencephalon/metabolism
14.
Elife ; 122024 Apr 24.
Article in English | MEDLINE | ID: mdl-38655918

ABSTRACT

Obstructive sleep apnea (OSA) is a prevalent sleep-related breathing disorder that results in multiple bouts of intermittent hypoxia. OSA has many neurological and systemic comorbidities, including dysphagia, or disordered swallow, and discoordination with breathing. However, the mechanism in which chronic intermittent hypoxia (CIH) causes dysphagia is unknown. Recently, we showed the postinspiratory complex (PiCo) acts as an interface between the swallow pattern generator (SPG) and the inspiratory rhythm generator, the preBötzinger complex, to regulate proper swallow-breathing coordination (Huff et al., 2023). PiCo is characterized by interneurons co-expressing transporters for glutamate (Vglut2) and acetylcholine (ChAT). Here we show that optogenetic stimulation of ChATcre:Ai32, Vglut2cre:Ai32, and ChATcre:Vglut2FlpO:ChR2 mice exposed to CIH does not alter swallow-breathing coordination, but unexpectedly disrupts swallow behavior via triggering variable swallow motor patterns. This suggests that glutamatergic-cholinergic neurons in PiCo are not only critical for the regulation of swallow-breathing coordination, but also play an important role in the modulation of swallow motor patterning. Our study also suggests that swallow disruption, as seen in OSA, involves central nervous mechanisms interfering with swallow motor patterning and laryngeal activation. These findings are crucial for understanding the mechanisms underlying dysphagia, both in OSA and other breathing and neurological disorders.


Subject(s)
Deglutition , Hypoxia , Animals , Mice , Deglutition/physiology , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Optogenetics , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Sleep Apnea, Obstructive/physiopathology , Sleep Apnea, Obstructive/metabolism , Cholinergic Neurons/physiology , Cholinergic Neurons/metabolism , Interneurons/physiology , Interneurons/metabolism , Respiration , Female
15.
Cell Rep ; 43(4): 114009, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38536818

ABSTRACT

To better understand the function of cholinergic projection neurons in the ventral pallidum (VP), we examined behavioral responses to appetitive (APP) and aversive (AV) odors that elicited approach or avoidance, respectively. Exposure to each odor increased cFos expression and calcium signaling in VP cholinergic neurons. Activity and Cre-dependent viral vectors selectively labeled VP cholinergic neurons that were activated and reactivated in response to either APP or AV odors, but not both, identifying two non-overlapping populations of VP cholinergic neurons differentially activated by the valence of olfactory stimuli. These two subpopulations showed differences in electrophysiological properties, morphology, and projections to the basolateral amygdala. Although VP neurons are engaged in both approach and avoidance behavioral responses, cholinergic signaling is only required for approach behavior. Thus, two distinct subpopulations of VP cholinergic neurons differentially encode valence of olfactory stimuli and play distinct roles in approach and avoidance behaviors.


Subject(s)
Basal Forebrain , Cholinergic Neurons , Odorants , Animals , Cholinergic Neurons/physiology , Basal Forebrain/physiology , Mice , Male , Smell/physiology , Mice, Inbred C57BL
16.
Cell Rep ; 43(4): 113999, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38554281

ABSTRACT

Motor neuron (MN) demise is a hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Post-transcriptional gene regulation can control RNA's fate, and defects in RNA processing are critical determinants of MN degeneration. N6-methyladenosine (m6A) is a post-transcriptional RNA modification that controls diverse aspects of RNA metabolism. To assess the m6A requirement in MNs, we depleted the m6A methyltransferase-like 3 (METTL3) in cells and mice. METTL3 depletion in embryonic stem cell-derived MNs has profound and selective effects on survival and neurite outgrowth. Mice with cholinergic neuron-specific METTL3 depletion display a progressive decline in motor behavior, accompanied by MN loss and muscle denervation, culminating in paralysis and death. Reader proteins convey m6A effects, and their silencing phenocopies METTL3 depletion. Among the m6A targets, we identified transactive response DNA-binding protein 43 (TDP-43) and discovered that its expression is under epitranscriptomic control. Thus, impaired m6A signaling disrupts MN homeostasis and triggers neurodegeneration conceivably through TDP-43 deregulation.


Subject(s)
Cholinergic Neurons , Methyltransferases , Neuromuscular Diseases , Animals , Humans , Mice , Adenosine/metabolism , Adenosine/analogs & derivatives , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/genetics , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Motor Neurons/metabolism , Motor Neurons/pathology , Neuromuscular Diseases/metabolism , Neuromuscular Diseases/pathology
17.
Neuron ; 112(8): 1342-1357.e6, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38359827

ABSTRACT

The basal forebrain (BF) is a complex structure that plays key roles in regulating various brain functions. However, it remains unclear how cholinergic and non-cholinergic BF neurons modulate large-scale functional networks and their relevance in intrinsic and extrinsic behaviors. With an optimized awake mouse optogenetic fMRI approach, we revealed that optogenetic stimulation of four BF neuron types evoked distinct cell-type-specific whole-brain BOLD activations, which could be attributed to BF-originated low-dimensional structural networks. Additionally, optogenetic activation of VGLUT2, ChAT, and PV neurons in the BF modulated the preference for locomotion, exploration, and grooming, respectively. Furthermore, we uncovered the functional network basis of the above BF-modulated behavioral preference through a decoding model linking the BF-modulated BOLD activation, low-dimensional structural networks, and behavioral preference. To summarize, we decoded the functional network basis of differential behavioral preferences with cell-type-specific optogenetic fMRI on the BF and provided an avenue for investigating mouse behaviors from a whole-brain view.


Subject(s)
Basal Forebrain , Animals , Mice , Basal Forebrain/physiology , Optogenetics , Magnetic Resonance Imaging , Neurons/physiology , Cholinergic Agents , Cholinergic Neurons/physiology
18.
Eur J Neurosci ; 59(7): 1638-1656, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38383047

ABSTRACT

Several brainstem nuclei degenerate in Parkinson's disease (PD). In addition to the well-characterized dopaminergic neurons of the substantia nigra pars compacta (SNc), the cholinergic neurons of the pedunculopontine nucleus (PPN) also degenerate in PD. One leading hypothesis of selective vulnerability is that pacemaking activity and the activation of low-threshold L-type calcium current are major contributors to tonic calcium load and cellular stress in SNc dopaminergic neurons. However, it is not yet clear whether the vulnerable PPN cholinergic neurons share this property. Therefore, we used two-photon dendritic calcium imaging and whole-cell electrophysiology to evaluate the role of L-type calcium channels in tonic and phasic dendritic calcium signals in PPN and SNc neurons. In addition, we investigated N- and P/Q-type calcium channel regulation of firing properties and dendritic calcium in PPN neurons. We found that blocking L-type channels reduces tonic firing rate and dendritic calcium levels in SNc neurons. By contrast, the tonic calcium load in PPN neurons did not depend on L-, N- or P/Q-type channels. However, we found that blocking either L-type (with nifedipine) or N- and P/Q-type (with omega-conotoxin MVIIC) channels reduces phasic calcium influx in PPN dendrites. Together, these findings show that L-type calcium channels play different roles in the activity of SNc and PPN neurons, and suggest that low-threshold L-type channels are not responsible for tonic calcium levels in PPN cholinergic neurons and are therefore not likely to be a source of selective vulnerability in these cells.


Subject(s)
Dopaminergic Neurons , Parkinson Disease , Humans , Calcium , Calcium Channels, L-Type , Substantia Nigra/physiology , Cholinergic Neurons , Cholinergic Agents
19.
J Comp Neurol ; 532(2): e25587, 2024 02.
Article in English | MEDLINE | ID: mdl-38335048

ABSTRACT

We examined the presence/absence and parcellation of cholinergic neurons in the hypothalami of five birds: a Congo grey parrot (Psittacus erithacus), a Timneh grey parrot (P. timneh), a pied crow (Corvus albus), a common ostrich (Struthio camelus), and an emu (Dromaius novaehollandiae). Using immunohistochemistry to an antibody raised against the enzyme choline acetyltransferase, hypothalamic cholinergic neurons were observed in six distinct clusters in the medial, lateral, and ventral hypothalamus in the parrots and crow, similar to prior observations made in the pigeon. The expression of cholinergic nuclei was most prominent in the Congo grey parrot, both in the medial and lateral hypothalamus. In contrast, no evidence of cholinergic neurons in the hypothalami of either the ostrich or emu was found. It is known that the expression of sleep states in the ostrich is unusual and resembles that observed in the monotremes that also lack hypothalamic cholinergic neurons. It has been proposed that the cholinergic system acts globally to produce and maintain brain states, such as those of arousal and rapid-eye-movement sleep. The hiatus in the cholinergic system of the ostrich, due to the lack of hypothalamic cholinergic neurons, may explain, in part, the unusual expression of sleep states in this species. These comparative anatomical and sleep studies provide supportive evidence for global cholinergic actions and may provide an important framework for our understanding of one broad function of the cholinergic system and possible dysfunctions associated with global cholinergic neural activity.


Subject(s)
Dromaiidae , Struthioniformes , Animals , Dromaiidae/metabolism , Struthioniformes/metabolism , Brain/metabolism , Hypothalamus/metabolism , Cholinergic Neurons/metabolism , Sleep/physiology , Cholinergic Agents , Choline O-Acetyltransferase/metabolism
20.
Neurosci Biobehav Rev ; 159: 105569, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38309497

ABSTRACT

Contextual and spatial systems facilitate changes in emotional memory regulation brought on by traumatic stress. Cholinergic basal forebrain (chBF) neurons provide input to contextual/spatial systems and although chBF neurons are important for emotional memory, it is unknown how they contribute to the traumatic stress effects on emotional memory. Clusters of chBF neurons that project to the prefrontal cortex (PFC) modulate fear conditioned suppression and passive avoidance, while clusters of chBF neurons that project to the hippocampus (Hipp) and PFC (i.e. cholinergic medial septum and diagonal bands of Broca (chMS/DBB neurons) are critical for fear extinction. Interestingly, neither Hipp nor PFC projecting chMS/DBB neurons are critical for fear extinction. The retrosplenial cortex (RSC) is a contextual/spatial memory system that receives input from chMS/DBB neurons, but whether this chMS/DBB-RSC circuit facilitates traumatic stress effects on emotional memory remain unexplored. Traumatic stress leads to neuroinflammation and the buildup of reactive oxygen species. These two molecular processes may converge to disrupt chBF circuits enhancing the impact of traumatic stress on emotional memory.


Subject(s)
Basal Forebrain , Extinction, Psychological , Humans , Extinction, Psychological/physiology , Fear/physiology , Hippocampus/physiology , Cholinergic Neurons
SELECTION OF CITATIONS
SEARCH DETAIL
...