Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 98
1.
Behav Brain Res ; 444: 114363, 2023 04 27.
Article En | MEDLINE | ID: mdl-36849047

BACKGROUND: Deep brain stimulation (DBS) is commonly used to alleviate motor symptoms in several movement disorders. However, the procedure is invasive, and the technology has remained largely stagnant since its inception decades ago. Recently, we have shown that wireless nanoelectrodes may offer an alternative approach to conventional DBS. However, this method is still in its infancy, and more research is required to characterize its potential before it can be considered as an alternative to conventional DBS. OBJECTIVES: Herein, we aimed to investigate the effect of stimulation via magnetoelectric nanoelectrodes on primary neurotransmitter systems that have implications for DBS in movement disorders. METHODS: Mice were injected with either magnetoelectric nanoparticles (MENPs) or magnetostrictive nanoparticles (MSNPs, as a control) in the subthalamic nucleus (STN). Mice then underwent magnetic stimulation, and their motor behavior was assessed in the open field test. In addition, magnetic stimulation was applied before sacrifice and post-mortem brains were processed for immunohistochemistry (IHC) to assess the co-expression of c-Fos with either tyrosine hydroxylase (TH), tryptophan hydroxylase-2 (TPH2) or choline acetyltransferase (ChAT). RESULTS: Stimulated animals covered longer distances in the open field test when compared to controls. Moreover, we found a significant increase in c-Fos expression in the motor cortex (MC) and paraventricular region of the thalamus (PV-thalamus) after magnetoelectric stimulation. Stimulated animals showed fewer TPH2/c-Fos double-labeled cells in the dorsal raphe nucleus (DRN), as well as TH/c-Fos double-labeled cells in the ventral tegmental area (VTA), but not in the substantia nigra pars compacta (SNc). There was no significant difference in the number of ChAT/ c-Fos double-labeled cells in the pedunculopontine nucleus (PPN). CONCLUSIONS: Magnetoelectric DBS in mice enables selective modulation of deep brain areas and animal behavior. The measured behavioral responses are associated with changes in relevant neurotransmitter systems. These changes are somewhat similar to those observed in conventional DBS, suggesting that magnetoelectric DBS might be a suitable alternative.


Deep Brain Stimulation , Movement Disorders , Pedunculopontine Tegmental Nucleus , Subthalamic Nucleus , Mice , Animals , Subthalamic Nucleus/metabolism , Deep Brain Stimulation/methods , Pedunculopontine Tegmental Nucleus/metabolism , Thalamus/metabolism , Proto-Oncogene Proteins c-fos/metabolism
2.
Neurosci Lett ; 802: 137134, 2023 04 01.
Article En | MEDLINE | ID: mdl-36801348

Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is standard care for severe motor symptoms of Parkinson's disease (PD). However, a challenge of DBS remains improving gait. Gait has been associated with the cholinergic system in the pedunculopontine nucleus (PPN). In this study, we investigated the effects of long-term intermittent bilateral STN-DBS on PPN cholinergic neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Parkinsonian mouse model. Motor behavior, previously assessed by the automated Catwalk gait analysis, demonstrated a parkinsonian-like motor phenotype with static and dynamic gait impairments, which were reversed by STN-DBS. In this study, a subset of brains was further immunohistochemically processed for choline acetyltransferase (ChAT) and the neuronal activation marker c-Fos. MPTP treatment resulted in a significant reduction of PPN ChAT expressing neurons compared to saline treatment. STN-DBS did not alter the number of ChAT expressing neurons, nor the number of double-labelled PPN neurons for ChAT and c-Fos. Although STN-DBS improved gait in our model this was not associated with an altered expression or activation of PPN acetylcholine neurons. Motor and gait effects of STN-DBS are therefore less likely to be mediated by the STN-PPN connection and PPN cholinergic system.


Deep Brain Stimulation , Pedunculopontine Tegmental Nucleus , Subthalamic Nucleus , Mice , Animals , Deep Brain Stimulation/methods , Pedunculopontine Tegmental Nucleus/metabolism , Cholinergic Neurons , Gait , Cholinergic Agents
3.
J Neural Transm (Vienna) ; 129(12): 1469-1479, 2022 12.
Article En | MEDLINE | ID: mdl-36222971

Prior studies indicate more severe brainstem cholinergic deficits in Progressive Supranuclear Palsy (PSP) compared to Parkinson's disease (PD), but the extent and topography of subcortical deficits remains poorly understood. The objective of this study is to investigate differential cholinergic systems changes in progressive supranuclear palsy (PSP, n = 8) versus Parkinson's disease (PD, n = 107) and older controls (n = 19) using vesicular acetylcholine transporter [18F]-fluoroethoxybenzovesamicol (FEOBV) positron emission tomography (PET). A whole-brain voxel-based PET analysis using Statistical Parametric Mapping (SPM) software (SPM12) for inter-group comparisons using parametric [18F]-FEOBV DVR images. Voxel-based analyses showed lower FEOBV binding in the tectum, metathalamus, epithalamus, pulvinar, bilateral frontal opercula, anterior insulae, superior temporal pole, anterior cingulum, some striatal subregions, lower brainstem, and cerebellum in PSP versus PD (p < 0.05; false discovery rate-corrected). More severe and diffuse reductions were present in PSP vs controls. Higher frequency of midbrain cholinergic losses was seen in PSP compared to the PD participants using 5th percentile normative cut-off values (χ2 = 4.12, p < 0.05). When compared to PD, these findings suggested disease-specific cholinergic vulnerability in the tectum, striatal cholinergic interneurons, and projections from the pedunculopontine nucleus, medial vestibular nucleus, and the cholinergic forebrain in PSP.


Parkinson Disease , Pedunculopontine Tegmental Nucleus , Supranuclear Palsy, Progressive , Humans , Supranuclear Palsy, Progressive/diagnostic imaging , Parkinson Disease/diagnostic imaging , Parkinson Disease/metabolism , Positron-Emission Tomography/methods , Pedunculopontine Tegmental Nucleus/metabolism , Cholinergic Agents
4.
Nat Commun ; 13(1): 504, 2022 01 26.
Article En | MEDLINE | ID: mdl-35082287

The pedunculopontine nucleus (PPN) is a locomotor command area containing glutamatergic neurons that control locomotor initiation and maintenance. These motor actions are deficient in Parkinson's disease (PD), where dopaminergic neurodegeneration alters basal ganglia activity. Being downstream of the basal ganglia, the PPN may be a suitable target for ameliorating parkinsonian motor symptoms. Here, we use in vivo cell-type specific PPN activation to restore motor function in two mouse models of parkinsonism made by acute pharmacological blockage of dopamine transmission. With a combination of chemo- and opto-genetics, we show that excitation of caudal glutamatergic PPN neurons can normalize the otherwise severe locomotor deficit in PD, whereas targeting the local GABAergic population only leads to recovery of slow locomotion. The motor rescue driven by glutamatergic PPN activation is independent of activity in nearby locomotor promoting glutamatergic Cuneiform neurons. Our observations point to caudal glutamatergic PPN neurons as a potential target for neuromodulatory restoration of locomotor function in PD.


Mesencephalon/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Parkinsonian Disorders/metabolism , Animals , Disease Models, Animal , Dopamine/metabolism , Excitatory Amino Acid Agents/pharmacology , Female , Male , Mesencephalon/drug effects , Mesencephalon/physiopathology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Parkinson Disease/physiopathology , Parkinsonian Disorders/physiopathology , Pedunculopontine Tegmental Nucleus/metabolism , Pedunculopontine Tegmental Nucleus/physiopathology
5.
Front Neural Circuits ; 15: 731333, 2021.
Article En | MEDLINE | ID: mdl-34675779

While electron microscopy represents the gold standard for detection of synapses, a number of limitations prevent its broad applicability. A key method for detecting synapses is immunostaining for markers of pre- and post-synaptic proteins, which can infer a synapse based upon the apposition of the two markers. While immunostaining and imaging techniques have improved to allow for identification of synapses in tissue, analysis and identification of these appositions are not facile, and there has been a lack of tools to accurately identify these appositions. Here, we delineate a macro that uses open-source and freely available ImageJ or FIJI for analysis of multichannel, z-stack confocal images. With use of a high magnification with a high NA objective, we outline two methods to identify puncta in either sparsely or densely labeled images. Puncta from each channel are used to eliminate non-apposed puncta and are subsequently linked with their cognate from the other channel. These methods are applied to analysis of a pre-synaptic marker, bassoon, with two different post-synaptic markers, gephyrin and N-methyl-d-aspartate (NMDA) receptor subunit 1 (NR1). Using gephyrin as an inhibitory, post-synaptic scaffolding protein, we identify inhibitory synapses in basolateral amygdala, central amygdala, arcuate and the ventromedial hypothalamus. Systematic variation of the settings identify the parameters most critical for this analysis. Identification of specifically overlapping puncta allows for correlation of morphometry data between each channel. Finally, we extend the analysis to only examine puncta overlapping with a cytoplasmic marker of specific cell types, a distinct advantage beyond electron microscopy. Bassoon puncta are restricted to virally transduced, pedunculopontine tegmental nucleus (PPN) axons expressing yellow fluorescent protein. NR1 puncta are restricted to tyrosine hydroxylase labeled dopaminergic neurons of the substantia nigra pars compacta (SNc). The macro identifies bassoon-NR1 overlap throughout the image, or those only restricted to the PPN-SNc connections. Thus, we have extended the available analysis tools that can be used to study synapses in situ. Our analysis code is freely available and open-source allowing for further innovation.


Pedunculopontine Tegmental Nucleus , Synapses , Dopaminergic Neurons/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , Receptors, N-Methyl-D-Aspartate , Synapses/metabolism , Tyrosine 3-Monooxygenase
6.
J Parkinsons Dis ; 11(4): 1773-1790, 2021.
Article En | MEDLINE | ID: mdl-34151857

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disorder associated with insoluble pathological aggregates of the protein α-synuclein. While PD is diagnosed by motor symptoms putatively due to aggregated α-synuclein-mediated damage to substantia nigra (SN) neurons, up to a decade before motor symptom appearance, patients exhibit sleep disorders (SDs). Therefore, we hypothesized that α-synuclein, which can be present in monomeric, fibril, and other forms, has deleterious cellular actions on sleep-control nuclei. OBJECTIVE: We investigated whether native monomer and fibril forms of α-synuclein have effects on neuronal function, calcium dynamics, and cell-death-induction in two sleep-controlling nuclei: the laterodorsal tegmentum (LDT), and the pedunculopontine tegmentum (PPT), as well as the motor-controlling SN. METHODS: Size exclusion chromatography, Thioflavin T fluorescence assays, and circular dichroism spectroscopy were used to isolate structurally defined forms of recombinant, human α-synuclein. Neuronal and viability effects of characterized monomeric and fibril forms of α-synuclein were determined on LDT, PPT, and SN neurons using electrophysiology, calcium imaging, and neurotoxicity assays. RESULTS: In LDT and PPT neurons, both forms of α-synuclein induced excitation and increased calcium, and the monomeric form heightened putatively excitotoxic neuronal death, whereas, in the SN, we saw inhibition, decreased intracellular calcium, and monomeric α-synuclein was not associated with heightened cell death. CONCLUSION: Nucleus-specific differential effects suggest mechanistic underpinnings of SDs' prodromal appearance in PD. While speculative, we hypothesize that the monomeric form of α-synuclein compromises functionality of sleep-control neurons, leading to the presence of SDs decades prior to motor dysfunction.


Parkinson Disease , Sleep Wake Disorders , alpha-Synuclein , Humans , Parkinson Disease/complications , Parkinson Disease/pathology , Pedunculopontine Tegmental Nucleus/metabolism , Sleep Wake Disorders/etiology , Substantia Nigra/metabolism , Tegmentum Mesencephali/metabolism , alpha-Synuclein/metabolism
7.
Front Neural Circuits ; 14: 43, 2020.
Article En | MEDLINE | ID: mdl-32765226

The inferior colliculus processes nearly all ascending auditory information. Most collicular cells respond to sound, and for a majority of these cells, the responses can be modulated by acetylcholine (ACh). The cholinergic effects are varied and, for the most part, the underlying mechanisms are unknown. The major source of cholinergic input to the inferior colliculus is the pedunculopontine tegmental nucleus (PPT), part of the pontomesencephalic tegmentum known for projections to the thalamus and roles in arousal and the sleep-wake cycle. Characterization of PPT inputs to the inferior colliculus has been complicated by the mixed neurotransmitter population within the PPT. Using selective viral-tract tracing techniques in a ChAT-Cre Long Evans rat, the present study characterizes the distribution and targets of cholinergic projections from PPT to the inferior colliculus. Following the deposit of viral vector in one PPT, cholinergic axons studded with boutons were present bilaterally in the inferior colliculus, with the greater density of axons and boutons ipsilateral to the injection site. On both sides, cholinergic axons were present throughout the inferior colliculus, distributing boutons to the central nucleus, lateral cortex, and dorsal cortex. In each inferior colliculus (IC) subdivision, the cholinergic PPT axons appear to contact both GABAergic and glutamatergic neurons. These findings suggest cholinergic projections from the PPT have a widespread influence over the IC, likely affecting many aspects of midbrain auditory processing. Moreover, the effects are likely to be mediated by direct cholinergic actions on both excitatory and inhibitory circuits in the inferior colliculus.


Cholinergic Neurons/metabolism , Inferior Colliculi/metabolism , Neurons/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , Animals , Axons/metabolism , Axons/pathology , Cholinergic Neurons/pathology , Inferior Colliculi/cytology , Inferior Colliculi/pathology , Neuroanatomical Tract-Tracing Techniques , Neurons/pathology , Pedunculopontine Tegmental Nucleus/cytology , Pedunculopontine Tegmental Nucleus/pathology , Rats , Rats, Long-Evans
8.
Neurotherapeutics ; 17(3): 1120-1141, 2020 07.
Article En | MEDLINE | ID: mdl-31965550

The brainstem-based pedunculopontine nucleus (PPN) traditionally associates with motor function, but undergoes extensive degeneration during Parkinson's disease (PD), which correlates with axial motor deficits. PPN-deep brain stimulation (DBS) can alleviate certain symptoms, but its mechanism(s) of action remains unknown. We previously characterized rats hemi-intranigrally injected with the proteasomal inhibitor lactacystin, as an accurate preclinical model of PD. Here we used a combination of chemogenetics with positron emission tomography imaging for in vivo interrogation of discrete neural networks in this rat model of PD. Stimulation of excitatory designer receptors exclusively activated by designer drugs expressed within PPN cholinergic neurons activated residual nigrostriatal dopaminergic neurons to produce profound motor recovery, which correlated with striatal dopamine efflux as well as restored dopamine receptor 1- and dopamine receptor 2-based medium spiny neuron activity, as was ascertained with c-Fos-based immunohistochemistry and stereological cell counts. By revealing that the improved axial-related motor functions seen in PD patients receiving PPN-DBS may be due to stimulation of remaining PPN cholinergic neurons interacting with dopaminergic ones in both the substantia nigra pars compacta and the striatum, our data strongly favor the PPN cholinergic-midbrain dopaminergic connectome as mechanism for PPN-DBS's therapeutic effects. These findings have implications for refining PPN-DBS as a promising treatment modality available to PD patients.


Cholinergic Neurons/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Parkinsonian Disorders/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , Animals , Cholinergic Neurons/drug effects , Corpus Striatum/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/genetics , Pedunculopontine Tegmental Nucleus/drug effects , Piperazines/pharmacology , Piperazines/therapeutic use , Rats , Rats, Long-Evans , Rats, Transgenic , Signal Transduction/drug effects , Signal Transduction/physiology
9.
Prog Neurobiol ; 185: 101734, 2020 02.
Article En | MEDLINE | ID: mdl-31863802

Prepulse inhibition (PPI) is an operational measure of sensorimotor gating. Deficits of PPI are a hallmark of schizophrenia and associated with several other psychiatric illnesses such as e.g. autism spectrum disorder, yet the mechanisms underlying PPI are still not fully understood. There is growing evidence contradicting the long-standing hypothesis that PPI is mediated by a short feed-forward midbrain circuitry including inhibitory cholinergic projections from the pedunculopontine tegmental nucleus (PPTg) to the startle pathway. Here, we employed a chemogenetic approach to explore the involvement of the PPTg in general, and cholinergic neurons specifically, in PPI. Activation of inhibitory DREADDs (designer receptors exclusively activated by designer drugs) in the PPTg by systemic administration of clozapine-N-oxide (CNO) disrupted PPI, confirming the involvement of the PPTg in PPI. In contrast, chemogenetic inhibition of specifically cholinergic PPTg neurons had no effect on PPI, but inhibited morphine-induced conditioned place preference (CPP) in the same animals, showing that the DREADDs were effective in modulating behavior. These findings support a functional role of the PPTg and/or neighboring structures in PPI in accordance with previous lesion studies, but also provide strong evidence against the hypothesis that specifically cholinergic PPTg neurons are involved in mediating PPI, implicating rather non-cholinergic midbrain neurons.


Autism Spectrum Disorder/metabolism , Mesencephalon/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , Prepulse Inhibition/physiology , Animals , Cholinergic Agents/metabolism , Female , Male , Rats, Long-Evans
10.
Am J Physiol Cell Physiol ; 318(2): C282-C288, 2020 02 01.
Article En | MEDLINE | ID: mdl-31747316

The pedunculopontine nucleus (PPN) is part of the reticular activating system (RAS) in charge of arousal and rapid eye movement sleep. The presence of high-frequency membrane oscillations in the gamma-band range in the PPN has been extensively demonstrated both in vivo and in vitro. Our group previously described histone deacetylation (HDAC) inhibition in vitro induced protein changes in F-actin cytoskeleton and intracellular Ca2+ concentration regulation proteins in the PPN. Here, we present evidence that supports the presence of a fine balance between HDAC function and calcium calmodulin kinase II-F-actin interactions in the PPN. We modified F-actin polymerization in vitro by using jasplakinolide (1 µM, a promoter of F-actin stabilization), or latrunculin-B (1 µM, an inhibitor of actin polymerization). Our results showed that shifting the balance in either direction significantly reduced PPN gamma oscillation as well as voltage-dependent calcium currents.


Actins/metabolism , Epigenesis, Genetic/physiology , Neurons/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , Actin Cytoskeleton/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Epigenesis, Genetic/genetics , Female , Male , Membrane Potentials/physiology , Rats , Rats, Sprague-Dawley
11.
Medicina (Kaunas) ; 55(10)2019 Sep 20.
Article En | MEDLINE | ID: mdl-31547185

Background and Objectives: The knowledge that the cholinergic neurons from pedunculopontine nucleus (PPN) are vulnerable to the degeneration in early stages of the Parkinson disease progression has opened new perspectives to the development of experimental model focused in pontine lesions that could increase the risk of nigral degeneration. In this context it is known that PPN lesioned rats exhibit early changes in the gene expression of proteins responsible for dopaminergic homeostasis. At the same time, it is known that nicotinic cholinergic receptors (nAChRs) mediate the excitatory influence of pontine-nigral projection. However, the effect of PPN injury on the expression of transcription factors that modulate dopaminergic neurotransmission in the adult brain as well as the α7 nAChRs gene expression has not been studied. The main objective of the present work was the study of the effects of the unilateral neurotoxic lesion of PPN in nuclear receptor-related factor 1 (Nurr1), paired-like homeodomain transcription factor 3 (Pitx3), and α7 nAChRs mRNA expression in nigral tissue. Materials and Methods: The molecular biology studies were performed by means of RT-PCR. The following experimental groups were organized: Non-treated rats, N-methyl-D-aspartate (NMDA)-lesioned rats, and Sham operated rats. Experimental subjects were sacrificed 24 h, 48 h and seven days after PPN lesion. Results: Nurr1 mRNA expression, showed a significant increase both 24 h (p < 0.001) and 48 h (p < 0.01) after PPN injury. Pitx3 mRNA expression evidenced a significant increase 24 h (p < 0.001) followed by a significant decrease 48 h and seven days after PPN lesion (p < 0.01). Finally, the α7 nAChRs nigral mRNA expression remained significantly diminished 24 h, 48 h (p < 0.001), and 7 days (p < 0.01) after PPN neurotoxic injury. Conclusion: Taking together these modifications could represent early warning signals and could be the preamble to nigral neurodegeneration events.


Dopaminergic Neurons/metabolism , Homeodomain Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Parkinson Disease/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , RNA, Messenger/metabolism , Substantia Nigra/metabolism , Transcription Factors/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Disease Models, Animal , Homeodomain Proteins/genetics , Male , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Parkinson Disease/pathology , Pedunculopontine Tegmental Nucleus/pathology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics
12.
eNeuro ; 6(4)2019.
Article En | MEDLINE | ID: mdl-31366590

Vesicular glutamate transporters (VGLUTs) mediate the synaptic uptake of glutamate from the cytosol into synaptic vesicles and are considered unambiguous neurochemical markers of glutamate neurons. However, many neurons not classically thought of as glutamatergic also express a VGLUT and co-release glutamate. Using a genetic fate-mapping strategy we found that most cholinergic neurons in the mouse mesopontine tegmentum express VGLUT2 at some point during development, including the pedunculopontine tegmental nucleus (PPTg), laterodorsal tegmental nucleus, and parabigeminal nucleus (PBG), but not the oculomotor nucleus. In contrast, very few of these cholinergic neurons displayed evidence of vesicular GABA transporter expression. Using multiplex fluorescent in situ hybridization, we determined that only PBG cholinergic neurons are also predominantly positive for VGLUT2 mRNA in the adult, with only small numbers of PPTg cholinergic neurons overlapping with VGLUT2 mRNA. Using Cre-dependent viral vectors we confirm these in situ hybridization data, and demonstrate projection patterns of cholinergic and glutamatergic populations. These results demonstrate that most mesopontine cholinergic neurons may transiently express VGLUT2, but that a large majority of PBG neurons retain VGLUT2 expression throughout adulthood, and support a growing body of literature indicating that distinct cholinergic populations have differing potential for GABA or glutamate co-release.


Cholinergic Neurons/metabolism , Mesencephalon/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Animals , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , RNA, Messenger/metabolism
13.
Neuropharmacology ; 158: 107744, 2019 11 01.
Article En | MEDLINE | ID: mdl-31437434

Cigarette smoking during pregnancy has been clinically associated with a variety of poorbehavioral outcomes for the exposed individuals, including higher risks for drug abuse and development of attention/deficit-hyperactive disorder (ADHD). Experimental studies support the hypothesis that nicotine might contribute to these risks, since prenatal nicotine exposure (PNE) in rodents was associated with greater addiction liability, hyperactivity, social impairments and a wide range of emotional and cognitive deficits. Alterations of glutamate signaling within brain regions involved in behavioral circuits could contribute to these outcomes. The pontine laterodorsal tegmental nucleus (LDT) exerts cholinergic modulation within the ventral tegmental area, nucleus accumbens, and cortical-projecting thalamic centers and PNE-associated alterations in LDT glutamate signaling could impact cholinergic output to these LDT targets. We have previously demonstrated that PNE alters AMPA-mediated signaling within LDT neurons, and in the present investigation, we focused on changes of NMDA receptors (NMDARs) and presence of silent synapses as an indicator of metaplastic processes in LDT cells associated with PNE treatment. PNE was associated with a decreased functional presence of GluN2B NMDAR subunits in synapses of large, putatively cholinergic neurons, whereas an increased function of this subunit was detected in small, likely GABAergic cells. In addition, PNE was associated with functional alterations of extrasynaptic NMDARs in putative cholinergic neurons, suggestive of an increased presence of GluN3A-containing NMDARs. An increased number of silent synapses was exclusively seen in the small cells. When taken together, we hypothesize that NMDA-mediated signaling changes within LDT neurons following PNE treatment would result in reductions of excitatory cholinergic modulatory tone in target brain regions, which would be expected to contribute to the behavioral deficits found among these individuals.


Cholinergic Neurons/drug effects , GABAergic Neurons/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Pedunculopontine Tegmental Nucleus/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Cholinergic Neurons/metabolism , Cigarette Smoking/metabolism , Female , GABAergic Neurons/metabolism , Mice , Patch-Clamp Techniques , Pedunculopontine Tegmental Nucleus/cytology , Pedunculopontine Tegmental Nucleus/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Synapses/drug effects , Synapses/metabolism
14.
Respir Physiol Neurobiol ; 269: 103254, 2019 11.
Article En | MEDLINE | ID: mdl-31325565

The pedunculopontine tegmental nucleus (PPTg) has been shown to have important functions relevant to the regulation of behavioral states and various motor control systems, including breathing control. The PPTg is considered an important nucleus in the mesopontine region with considerably cholinergic input to the ventral respiratory column. In addition, recent studies indicate that cholinergic innervation of the ventral respiratory column may play an important role in modulation of breathing. Here, we investigated the cholinergic stimulation of the PPTg and the changes in breathing output in conscious rats. Male Wistar rats (280-350 g, N = 5-12/group) with unilateral stainless steel cannula implanted into the PPTg were used. Respiratory parameters (tidal volume (VT), respiratory frequency (fR) and ventilation (VE)) were analyzed by whole body plethysmography. In unrestrained awake rats, unilateral injection of the cholinergic muscarinic agonist carbachol (10 mM-100 nL) in the PPTg decreased fR, and increase VT, without changing VE. The changes in fR and VT elicited by carbachol into the PPTg are abolished by previous blockade of the M4 muscarinic cholinergic receptors tropicamide into the PPTg. No significant changes in fR and VT elicited by carbachol were observed after blockade of the M1 and/or M3 muscarinic cholinergic receptors pirenzepine or 4-DAMP into the PPTg. Our data suggest that the changes in fR and VT produced by muscarinic cholinergic stimulation of PPTg is presumably mediated through a Gi-coupled M4 muscarinic receptors.


Pedunculopontine Tegmental Nucleus/metabolism , Receptor, Muscarinic M4/metabolism , Respiration/radiation effects , Animals , Consciousness , Male , Rats , Rats, Wistar
15.
Brain Res ; 1715: 134-147, 2019 07 15.
Article En | MEDLINE | ID: mdl-30914249

The mesolimbic system, particularly the somatodendritic ventral tegmental area (VTA), is responsible for the positive reinforcing aspects of various homeostatic stimuli. In turn, the pedunculopontine tegmental nucleus (PPN) is anatomically and functionally connected with the VTA and substantia nigra (SN). In the present study, we investigated the role of glutamate receptors in the PPN in motivated behaviors by using a model of feeding induced by electrical stimulation of the VTA in male Wistar rats (n = 80). We found that injection of 2.5/5 µg dizocilpine (MK-801; NMDA receptor antagonist) to the PPN significantly reduced the feeding response induced by unilateral VTA-stimulation. This reaction was significantly impaired after local injection of MK-801 into the PPN in the ipsilateral rather than the contralateral hemisphere. After NMDA injection (2/3 µg) to the PPN we did not observe behavioral changes, only a trend of a lengthening/shortening of the latency to a feeding reaction at the highest dose of NMDA (3 µg). Immunohistochemical TH+/c-Fos+ analysis revealed a decrease in the number of TH+ cells in the midbrain (VTA-SN) in all experimental groups and altered activity of c-Fos+ neurons in selected brain structures depending on drug type (MK-801/NMDA) and injection site (ipsi-/contralateral hemisphere). Additionally, the pattern of TH+/c-Fos+ expression showed lateralization of feeding circuit functional connectivity. We conclude that the level of NMDA receptor arousal in the PPN regulates the activity of the midbrain dopaminergic cells, and the PPN-VTA circuit may be important in the regulation of motivational aspects of food intake.


Feeding Behavior/physiology , Pedunculopontine Tegmental Nucleus/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Behavior, Animal/drug effects , Brain/drug effects , Dizocilpine Maleate/pharmacology , Dopaminergic Neurons/physiology , Male , Mesencephalon/physiology , Motivation/drug effects , N-Methylaspartate/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/physiology , Substantia Nigra/drug effects , Ventral Tegmental Area/drug effects
16.
Brain Res Bull ; 146: 164-170, 2019 03.
Article En | MEDLINE | ID: mdl-30634015

Electrical stimulation of the anterior pretectal nucleus (APtN) activates two descending pain inhibitory pathways. One of these pathways relays in the ipsilateral lateral paragigantocellular nucleus (LPGi), whereas the other pathway relays in the contralateral pedunculopontine tegmental nucleus (PPTg). Antinociceptive effect of APtN stimulation has been seen in various pain models in the rodents. Similarly, LPGi or PPTg stimulation results in higher pain thresholds. Descending antinociceptive pathways activated by electrical APtN stimulation have been elucidated, but the underlying neurotransmitter mechanisms involved have not been clarified yet. This study investigates the role that endogenous signaling plays in the ipsilateral LPGi or contralateral PPTg after the APtN is stimulated in the tail-flick test. First, we submitted rats to excitotoxic injection of N-methyl-d-aspartate (NMDA) into the contralateral PPTg. Then, we examined whether blockage of NMDA (AP-7), serotonergic (methysergide), or opioid (naloxone) receptors in the ipsilateral LPGi is required for APtN stimulation-evoked analgesia (SEA). Likewise, we examined the effects of antagonists of NMDA, serotonergic, or cholinergic nicotinic (mecamylamine) receptors on the contralateral PPTg in ipsilateral LPGi-lesioned rats. Our results confirmed that APtN stimulation activates two pain inhibitory pathways and showed that endogenous opioid signaling in the ipsilateral LPGi appears to be necessary for APtN SEA and for endogenous NMDA, serotoninergic, and nicotinergic signaling in the contralateral PPTg.


Pain Management/methods , Pain/metabolism , Pretectal Region/drug effects , Analgesia/methods , Analgesics, Opioid , Animals , Electric Stimulation , Male , Methysergide , N-Methylaspartate/pharmacology , Naloxone , Narcotic Antagonists/pharmacology , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/pharmacology , Nociceptors/metabolism , Pain/physiopathology , Pain Measurement/drug effects , Pedunculopontine Tegmental Nucleus/drug effects , Pedunculopontine Tegmental Nucleus/metabolism , Pretectal Region/metabolism , Rats , Rats, Wistar
17.
Neurobiol Dis ; 128: 9-18, 2019 08.
Article En | MEDLINE | ID: mdl-30149181

The pedunculopontine nucleus (PPN) is part of the mesencephalic locomotor region (MLR) and has been involved in the control of gait, posture, locomotion, sleep, and arousal. It likely participates in some motor and non-motor symptoms of Parkinson's disease and is regularly proposed as a surgical target to ameliorate gait, posture and sleep disorders in Parkinsonian patients. The PPN overlaps with the monoaminergic systems including dopamine, serotonin and noradrenaline in the modulation of the above-mentioned functions. All these systems are involved in Parkinson's disease and the mechanism of the anti-Parkinsonian agents, mostly L-DOPA. This suggests that PPN interacts with monoaminergic neurons and vice versa. Some evidence indicates that the PPN sends cholinergic, glutamatergic and even gabaergic inputs to mesencephalic dopaminergic cells, with the data regarding serotonergic or noradrenergic cells being less well known. Similarly, the control exerted by the PPN on dopaminergic neurons, is multiple and complex, and more extensively explored than the other monoaminergic systems. The data on the influence of monoaminergic systems on PPN neuron activity are rather scarce. While there is evidence that the PPN influences the therapeutic response of L-DOPA, it is still difficult to discerne the reciprocal action of the PPN and monoaminergic systems in this action. Additional data are required to better understand the functional organization of monoaminergic inputs to the MLR including the PPN to get a clearer picture of their interaction.


Adrenergic Neurons/physiology , Antiparkinson Agents/therapeutic use , Dopaminergic Neurons/physiology , Levodopa/therapeutic use , Parkinson Disease/drug therapy , Pedunculopontine Tegmental Nucleus/metabolism , Serotonergic Neurons/physiology , Animals , Antiparkinson Agents/pharmacology , Humans , Levodopa/pharmacology , Parkinson Disease/metabolism , Pedunculopontine Tegmental Nucleus/drug effects
18.
Bipolar Disord ; 21(2): 108-116, 2019 03.
Article En | MEDLINE | ID: mdl-30506611

OBJECTIVES: This limited review examines the role of the reticular activating system (RAS), especially the pedunculopontine nucleus (PPN), one site of origin of bottom-up gamma, in the symptoms of bipolar disorder (BD). METHODS: The expression of neuronal calcium sensor protein 1 (NCS-1) in the brains of BD patients is increased. It has recently been found that all PPN neurons manifest intrinsic membrane beta/gamma frequency oscillations mediated by high threshold calcium channels, suggesting that it is one source of bottom-up gamma. This review specifically addresses the involvement of these channels in the manifestation of BD. RESULTS: Excess NCS-1 was found to dampen gamma band oscillations in PPN neurons. Lithium, a first line treatment for BD, was found to decrease the effects of NCS-1 on gamma band oscillations in PPN neurons. Moreover, gamma band oscillations appear to epigenetically modulate gene transcription in PPN neurons, providing a new direction for research in BD. CONCLUSIONS: This is an area needing much additional research, especially since the dysregulation of calcium channels may help explain many of the disorders of arousal in, elicit unwanted neuroepigenetic modulation in, and point to novel therapeutic avenues for, BD.


Bipolar Disorder/metabolism , Gamma Rhythm/physiology , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/metabolism , Pedunculopontine Tegmental Nucleus/metabolism , Animals , Bipolar Disorder/genetics , Bipolar Disorder/pathology , Calcium Channels/metabolism , Epigenesis, Genetic , Humans , Neurons/metabolism , Neurons/pathology
19.
Sci Rep ; 8(1): 13156, 2018 09 03.
Article En | MEDLINE | ID: mdl-30177751

Epigenetic mechanisms (i.e., histone post-translational modification and DNA methylation) play a role in regulation of gene expression. The pedunculopontine nucleus (PPN), part of the reticular activating system, manifests intrinsic gamma oscillations generated by voltage-dependent, high threshold N- and P/Q-type Ca2+ channels. We studied whether PPN intrinsic gamma oscillations are affected by inhibition of histone deacetylation. We showed that, a) acute in vitro exposure to the histone deacetylation Class I and II inhibitor trichostatin A (TSA, 1 µM) eliminated oscillations in the gamma range, but not lower frequencies, b) pre-incubation with TSA (1 µM, 90-120 min) also decreased gamma oscillations, c) Ca2+ currents (ICa) were reduced by TSA, especially on cells with P/Q-type channels, d) a HDAC Class I inhibitor MS275 (500 nM), and a Class IIb inhibitor Tubastatin A (150-500 nM), failed to affect gamma oscillations, e) MC1568, a HDAC Class IIa inhibitor (1 µM), blocked gamma oscillations, and f) the effects of both TSA and MC1568 were blunted by blockade of CaMKII with KN-93 (1 µM). These results suggest a cell type specific effect on gamma oscillations when histone deacetylation is blocked, suggesting that gamma oscillations through P/Q-type channels modulated by CaMKII may be linked to processes related to gene transcription.


Calcium Channels, P-Type/genetics , Calcium Channels, Q-Type/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Neurons/drug effects , Pedunculopontine Tegmental Nucleus/drug effects , Animals , Animals, Newborn , Benzamides/pharmacology , Benzylamines/pharmacology , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Gamma Rhythm/drug effects , Gamma Rhythm/physiology , Gene Expression Regulation , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Microtomy , Neurons/cytology , Neurons/metabolism , Pedunculopontine Tegmental Nucleus/cytology , Pedunculopontine Tegmental Nucleus/metabolism , Primary Cell Culture , Pyridines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Sulfonamides/pharmacology , Tissue Culture Techniques , Transcription, Genetic
20.
Mov Disord ; 33(5): 827-834, 2018 05.
Article En | MEDLINE | ID: mdl-29508906

BACKGROUND: The etiology of cervical dystonia is unknown. Cholinergic abnormalities have been identified in dystonia animal models and human imaging studies. Some animal models have cholinergic neuronal loss in the striatum and increased acetylcholinesterase activity in the pedunculopontine nucleus. OBJECTIVES: The objective of this study was to determine the presence of cholinergic abnormalities in the putamen and pedunculopontine nucleus in cervical dystonia human brain donors. METHODS: Formalin-fixed brain tissues were obtained from 8 cervical dystonia and 7 age-matched control brains (controls). Pedunculopontine nucleus was available in only 6 cervical dystonia and 5 controls. Neurodegeneration was evaluated pathologically in the putamen, pedunculopontine nucleus, and other regions. Cholinergic neurons were detected using choline acetyltransferase immunohistochemistry in the putamen and pedunculopontine nucleus. Putaminal cholinergic neurons were quantified. A total of 6 cervical dystonia patients and 6 age-matched healthy controls underwent diffusion tensor imaging to determine if there were white matter microstructural abnormalities around the pedunculopontine nucleus. RESULTS: Decreased or absent choline acetyltransferase staining was identified in all 6 pedunculopontine nucleus samples in cervical dystonia. In contrast, strong choline acetyltransferase staining was present in 4 of 5 pedunculopontine nucleus controls. There were no differences in pedunculopontine nucleus diffusion tensor imaging between cervical dystonia and healthy controls. There was no difference in numbers of putaminal cholinergic neurons between cervical dystonia and controls. CONCLUSIONS: Our findings suggest that pedunculopontine nucleus choline acetyltransferase deficiency represents a functional cholinergic deficit in cervical dystonia. Structural lesions and confounding neurodegenerative processes were excluded by absence of neuronal loss, gliosis, diffusion tensor imaging abnormalities, and beta-amyloid, tau, and alpha-synuclein pathologies. © 2018 International Parkinson and Movement Disorder Society.


Choline O-Acetyltransferase/deficiency , Cholinergic Neurons/pathology , Pedunculopontine Tegmental Nucleus/metabolism , Torticollis/pathology , Acetylcholine , Aged , Aged, 80 and over , Cholinergic Neurons/metabolism , Diffusion Tensor Imaging , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Pedunculopontine Tegmental Nucleus/diagnostic imaging , Torticollis/diagnostic imaging , Ubiquitin , tau Proteins/metabolism
...