Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
1.
J Acoust Soc Am ; 155(5): 3183-3194, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38738939

ABSTRACT

Medial olivocochlear (MOC) efferents modulate outer hair cell motility through specialized nicotinic acetylcholine receptors to support encoding of signals in noise. Transgenic mice lacking the alpha9 subunits of these receptors (α9KOs) have normal hearing in quiet and noise, but lack classic cochlear suppression effects and show abnormal temporal, spectral, and spatial processing. Mice deficient for both the alpha9 and alpha10 receptor subunits (α9α10KOs) may exhibit more severe MOC-related phenotypes. Like α9KOs, α9α10KOs have normal auditory brainstem response (ABR) thresholds and weak MOC reflexes. Here, we further characterized auditory function in α9α10KO mice. Wild-type (WT) and α9α10KO mice had similar ABR thresholds and acoustic startle response amplitudes in quiet and noise, and similar frequency and intensity difference sensitivity. α9α10KO mice had larger ABR Wave I amplitudes than WTs in quiet and noise. Other ABR metrics of hearing-in-noise function yielded conflicting findings regarding α9α10KO susceptibility to masking effects. α9α10KO mice also had larger startle amplitudes in tone backgrounds than WTs. Overall, α9α10KO mice had grossly normal auditory function in quiet and noise, although their larger ABR amplitudes and hyperreactive startles suggest some auditory processing abnormalities. These findings contribute to the growing literature showing mixed effects of MOC dysfunction on hearing.


Subject(s)
Acoustic Stimulation , Behavior, Animal , Noise , Animals , Female , Male , Mice , Auditory Pathways/physiology , Auditory Pathways/physiopathology , Auditory Perception/physiology , Auditory Threshold , Cochlea/physiology , Cochlea/physiopathology , Evoked Potentials, Auditory, Brain Stem , Hearing , Mice, Inbred C57BL , Mice, Knockout , Noise/adverse effects , Olivary Nucleus/physiology , Perceptual Masking , Phenotype , Receptors, Nicotinic/genetics , Receptors, Nicotinic/deficiency , Reflex, Startle
2.
Nat Commun ; 12(1): 2449, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33907194

ABSTRACT

In the developing auditory system, spontaneous activity generated in the cochleae propagates into the central nervous system to promote circuit formation. The effects of peripheral firing patterns on spontaneous activity in the central auditory system are not well understood. Here, we describe wide-spread bilateral coupling of spontaneous activity that coincides with the period of transient efferent modulation of inner hair cells from the brainstem medial olivocochlear system. Knocking out α9/α10 nicotinic acetylcholine receptors, a requisite part of the efferent pathway, profoundly reduces bilateral correlations. Pharmacological and chemogenetic experiments confirm that the efferent system is necessary for normal bilateral coupling. Moreover, auditory sensitivity at hearing onset is reduced in the absence of pre-hearing efferent modulation. Together, these results demonstrate how afferent and efferent pathways collectively shape spontaneous activity patterns and reveal the important role of efferents in coordinating bilateral spontaneous activity and the emergence of functional responses during the prehearing period.


Subject(s)
Auditory Pathways/physiology , Cochlea/physiology , Efferent Pathways/physiology , Feedback, Physiological , Receptors, Nicotinic/genetics , Acoustic Stimulation , Animals , Auditory Pathways/cytology , Cochlea/cytology , Functional Laterality/physiology , Gene Expression , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Inner/physiology , Inferior Colliculi/cytology , Inferior Colliculi/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Olivary Nucleus/cytology , Olivary Nucleus/physiology , Receptors, Nicotinic/deficiency
3.
Nature ; 581(7807): 204-208, 2020 05.
Article in English | MEDLINE | ID: mdl-32405000

ABSTRACT

It has been speculated that brain activities might directly control adaptive immune responses in lymphoid organs, although there is little evidence for this. Here we show that splenic denervation in mice specifically compromises the formation of plasma cells during a T cell-dependent but not T cell-independent immune response. Splenic nerve activity enhances plasma cell production in a manner that requires B-cell responsiveness to acetylcholine mediated by the α9 nicotinic receptor, and T cells that express choline acetyl transferase1,2 probably act as a relay between the noradrenergic nerve and acetylcholine-responding B cells. We show that neurons in the central nucleus of the amygdala (CeA) and the paraventricular nucleus (PVN) that express corticotropin-releasing hormone (CRH) are connected to the splenic nerve; ablation or pharmacogenetic inhibition of these neurons reduces plasma cell formation, whereas pharmacogenetic activation of these neurons increases plasma cell abundance after immunization. In a newly developed behaviour regimen, mice are made to stand on an elevated platform, leading to activation of CeA and PVN CRH neurons and increased plasma cell formation. In immunized mice, the elevated platform regimen induces an increase in antigen-specific IgG antibodies in a manner that depends on CRH neurons in the CeA and PVN, an intact splenic nerve, and B cell expression of the α9 acetylcholine receptor. By identifying a specific brain-spleen neural connection that autonomically enhances humoral responses and demonstrating immune stimulation by a bodily behaviour, our study reveals brain control of adaptive immunity and suggests the possibility to enhance immunocompetency by behavioural intervention.


Subject(s)
Behavior, Animal/physiology , Brain/physiology , Immunity, Humoral/immunology , Spleen/immunology , Spleen/innervation , Acetylcholine/metabolism , Acetylcholine/pharmacology , Adrenergic Neurons/metabolism , Amygdala/cytology , Amygdala/drug effects , Amygdala/metabolism , Animals , Brain/cytology , Brain/drug effects , Choline O-Acetyltransferase/metabolism , Corticotropin-Releasing Hormone/metabolism , Hemocyanins/immunology , Immunoglobulin G/immunology , Lymphocyte Activation , Male , Mice , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Plasma Cells/cytology , Plasma Cells/drug effects , Plasma Cells/immunology , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/metabolism , Spleen/cytology , Spleen/drug effects , Stress, Psychological/immunology , Stress, Psychological/metabolism , T-Lymphocytes/immunology
4.
Sci Adv ; 6(15): eaax8382, 2020 04.
Article in English | MEDLINE | ID: mdl-32284992

ABSTRACT

For decades, numerous studies have proposed that fast muscles contribute to quick movement, while slow muscles underlie locomotion requiring endurance. By generating mutant zebrafish whose fast muscles are synaptically silenced, we examined the contribution of fast muscles in both larval and adult zebrafish. In the larval stage, mutants lacked the characteristic startle response to tactile stimuli: bending of the trunk (C-bend) followed by robust forward propulsion. Unexpectedly, adult mutants with silenced fast muscles showed robust C-bends and forward propulsion upon stimulation. Retrograde labeling revealed that motor neurons genetically programmed to form synapses on fast muscles are instead rerouted and innervate slow muscles, which led to partial conversion of slow and intermediate muscles to fast muscles. Thus, extended silencing of fast muscle synapses changed motor neuron innervation and caused muscle cell type conversion, revealing an unexpected mechanism of locomotory adaptation.


Subject(s)
Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Neuromuscular Junction/physiology , Animals , Animals, Genetically Modified , Gene Knockdown Techniques , Gene Targeting , Genetic Loci , Larva , Locomotion , Receptors, Nicotinic/deficiency , Synaptic Transmission , Zebrafish
5.
Int Immunopharmacol ; 84: 106496, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32304995

ABSTRACT

Mucociliary clearance, the continuous removal of mucus-trapped particles by cilia-driven directed transport of the airway lining fluid, is the primary innate defense mechanism of the airways. It is potently activated by acetylcholine (ACh) addressing muscarinic receptors with a currently less defined role of nicotinic ACh receptors (nAChR). We here set out to determine their contribution in driving ciliary activity in an explanted mouse trachea preparation utilizing selected agonists and antagonists and nAChR-subunit deficient mice. Nicotine (100 µM) induced an increase in ciliary beat frequency, accompanied by a sharp, but not long lasting increase in particle transport speed (PTS) on the mucosal surface showing marked desensitization within the next 30 min. Nicotine-induced PTS acceleration was sensitive to the general nAChR inhibitors mecamylamine and d-tubocurarine as well as to the α3ß4-nAChR antagonist α-conotoxin AulB, but not to other antagonists primarily addressing α3ß2-nAChR or α4-, α7- and α9-containing nAChR. Agonists at α3ß*-nAChR (epibatidine, cytisine), but not cotinine mimicked the effect. Tracheas from mice with genetic deletion of nAChR subunits α5, α7, α9, α10, α9/10, and ß2 retained full PTS response to nicotine, whereas this was entirely lost in tracheas from mice lacking the ß4-subunit. Collectively, our data show that nicotinic stimulation of α3ß4-nAChR acutely increases PTS to the same extent as the established strong activator ATP. In view of the marked desensitization observed in the present setting, the physiological relevance of these receptors in adapting mucociliary clearance to rapidly changing endogenous or environmental stimuli remains open.


Subject(s)
Cilia/drug effects , Cilia/metabolism , Movement/drug effects , Nicotine/pharmacology , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Trachea/drug effects , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Protein Subunits/physiology , Receptors, Nicotinic/deficiency
6.
Mar Drugs ; 18(4)2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32272701

ABSTRACT

The α9-containing nicotinic acetylcholine receptor (nAChR) is increasingly emerging as a new tumor target owing to its high expression specificity in breast cancer. αO-Conotoxin GeXIVA is a potent antagonist of α9α10 nAChR. Nevertheless, the anti-tumor effect of GeXIVA on breast cancer cells remains unclear. Cell Counting Kit-8 assay was used to study the cell viability of breast cancer MDA-MD-157 cells and human normal breast epithelial cells, which were exposed to different doses of GeXIVA. Flow cytometry was adopted to detect the cell cycle arrest and apoptosis of GeXIVA in breast cancer cells. Migration ability was analyzed by wound healing assay. Western blot (WB), quantitative real-time PCR (QRT-PCR) and flow cytometry were used to determine expression of α9-nAChR. Stable MDA-MB-157 breast cancer cell line, with the α9-nAChR subunit knocked out (KO), was established using the CRISPR/Cas9 technique. GeXIVA was able to significantly inhibit the proliferation and promote apoptosis of breast cancer MDA-MB-157 cells. Furthermore, the proliferation of breast cancer MDA-MB-157 cells was inhibited by GeXIVA, which caused cell cycle arrest through downregulating α9-nAChR. GeXIVA could suppress MDA-MB-157 cell migration as well. This demonstrates that GeXIVA induced a downregulation of α9-nAChR expression, and the growth of MDA-MB-157 α9-nAChR KO cell line was inhibited as well, due to α9-nAChR deletion. GeXIVA inhibits the growth of breast cancer cell MDA-MB-157 cells in vitro and may occur in a mechanism abolishing α9-nAChR.


Subject(s)
Cell Proliferation/drug effects , Conotoxins/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/drug effects , Breast Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Female , Humans , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/metabolism
7.
Neuropharmacology ; 166: 107921, 2020 04.
Article in English | MEDLINE | ID: mdl-31881170

ABSTRACT

Desensitization of the nicotinic acetylcholine receptor (nAChR) containing the ß2 subunit is a potentially critical mechanism underlying the body weight (BW) reducing effects of nicotine. The purpose of this study was a) to determine the α subunit(s) that partners with the ß2 subunit to form the nAChR subtype that endogenously regulates energy balance and b) to probe the extent to which nAChR desensitization could be involved in the regulation of BW. We demonstrate that deletion of either the α4 or the ß2, but not the α5, subunit of the nAChR suppresses weight gain in a sex-dependent manner. Furthermore, chronic treatment with the ß2-selective nAChR competitive antagonist dihydro-ß-erythroidine (DHßE) in mice fed a high-fat diet suppresses weight gain. These results indicate that heteromeric α4ß2 nAChRs play a role as intrinsic regulators of energy balance and that desensitizing or inhibiting this nAChR is likely a relevant mechanism and thus could be a strategy for weight loss.


Subject(s)
Body Weight/physiology , Dihydro-beta-Erythroidine/administration & dosage , Receptors, Nicotinic/metabolism , Animals , Body Weight/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Diet, High-Fat/adverse effects , Female , Infusion Pumps , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Nicotinic/deficiency , Weight Gain/drug effects , Weight Gain/physiology
8.
Physiol Rep ; 7(6): e14023, 2019 03.
Article in English | MEDLINE | ID: mdl-30891952

ABSTRACT

Our previous immunoprecipitation analysis of nicotinic acetylcholine receptors (nAChRs) in the mouse superior cervical ganglion (SCG) revealed that approximately 55%, 24%, and 21% of receptors are comprised of α3ß4, α3ß4α5, and α3ß4ß2 subunits, respectively. Moreover, mice lacking ß4 subunits do not express α5-containing receptors but still express a small number of α3ß2 receptors. Here, we investigated how synaptic transmission is affected in the SCG of α5ß4-KO and α5ß2-KO mice. Using an ex vivo SCG preparation, we stimulated the preganglionic cervical sympathetic trunk and measured compound action potentials (CAPs) in the postganglionic internal carotid nerve. We found that CAP amplitude was unaffected in α5ß4-KO and α5ß2-KO ganglia, whereas the stimulation threshold for eliciting CAPs was significantly higher in α5ß4-KO ganglia. Moreover, intracellular recordings in SCG neurons revealed no difference in EPSP amplitude. We also found that the ganglionic blocking agent hexamethonium was the most potent in α5ß4-KO ganglia (IC50 : 22.1 µmol/L), followed by α5ß2-KO (IC50 : 126.7 µmol/L) and WT ganglia (IC50 : 389.2 µmol/L). Based on these data, we estimated an IC50 of 568.6 µmol/L for a receptor population consisting solely of α3ß4α5 receptors; and we estimated that α3ß4α5 receptors comprise 72% of nAChRs expressed in the mouse SCG. Similarly, by measuring the effects of hexamethonium on ACh-induced currents in cultured SCG neurons, we found that α3ß4α5 receptors comprise 63% of nAChRs. Thus, in contrast to our results obtained using immunoprecipitation, these data indicate that the majority of receptors at the cell surface of SCG neurons consist of α3ß4α5.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Nicotinic/metabolism , Superior Cervical Ganglion/metabolism , Synaptic Transmission , Animals , Cells, Cultured , Ganglionic Blockers/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Superior Cervical Ganglion/drug effects , Synaptic Potentials , Synaptic Transmission/drug effects
9.
Bone ; 120: 285-296, 2019 03.
Article in English | MEDLINE | ID: mdl-30414510

ABSTRACT

Alterations in bone strength and structure were found in knockout (KO) mouse strains with deletion of several acetylcholine receptors. Interestingly, the expression of the nicotinic acetylcholine receptors (nAChR) subunit α10 was down-regulated in osteogenic differentiated mesenchymal stem cells of patients with osteoporosis whereas the expression of subunit α9 was not altered. Since nAChR subunits α9 and α10 are often combined in a functional receptor, we analyzed here the bone of adult female KO mice with single deletion of either nAChR alpha9 (α9KO) or alpha10 (α10KO). Biomechanical testing showed a significant decrease of bending stiffness and maximal breaking force in α9KO compared to their corresponding wild type mice. Furthermore, an increase in trabecular pattern factor (Tb.Pf) and structure model index (SMI) was detected by µCT in α9KO indicating reduced bone mass. On the mRNA level a decrease of Collagen 1α1 and Connexin-43 was measured by real-time RT-PCR in α9KO while no alteration of osteoclast markers was detected in either mouse strain. Using electron microcopy we observed an increase in the number of osteocytes that showed signs of degeneration and cell death in the α9KO compared to their wild type mice, while α10KO showed no differences. In conclusion, we demonstrate alterations in bone strength, structure and bio-marker expression in α9KO mice which imply the induction of osteocyte degeneration. Thus, our data suggest that nAChR containing the α9 subunit might be involved in the homeostasis of osteocytes and therefore in bone mass regulation.


Subject(s)
Bone and Bones/anatomy & histology , Gene Deletion , Receptors, Nicotinic/genetics , Alkaline Phosphatase/metabolism , Animals , Animals, Newborn , Bone and Bones/physiology , Cancellous Bone/anatomy & histology , Cortical Bone/anatomy & histology , Female , Femur/anatomy & histology , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteocytes/metabolism , Osteocytes/ultrastructure , Receptors, Nicotinic/deficiency
10.
PLoS One ; 13(12): e0208982, 2018.
Article in English | MEDLINE | ID: mdl-30543688

ABSTRACT

Cholinergic Receptor Nicotinic Alpha 5 (CHRNA5) is an important susceptibility locus for nicotine addiction and lung cancer. Depletion of CHRNA5 has been associated with reduced cell viability, increased apoptosis and alterations in cellular motility in different cancers yet not in breast cancer. Herein we first showed the expression of CHRNA5 was variable and positively correlated with the fraction of total genomic alterations in breast cancer cell lines and tumors indicating its potential role in DNA damage response (DDR). Next, we demonstrated that silencing of CHRNA5 expression in MCF7 breast cancer cell line by RNAi affected expression of genes involved in cytoskeleton, TP53 signaling, DNA synthesis and repair, cell cycle, and apoptosis. The transcription profile of CHRNA5 depleted MCF7 cells showed a significant positive correlation with that of A549 lung cancer cell line while exhibiting a negative association with the CHRNA5 co-expression profile obtained from Cancer Cell Line Encylopedia (CCLE). Moreover, it exhibited high similarities with published MCF7 expression profiles obtained from exposure to TP53 inducer nutlin-3a and topoisomerase inhibitors. We then demonstrated that CHRNA5 siRNA treatment reduced cell viability and DNA synthesis indicating G1 arrest while it significantly increased apoptotic sub-G1 cell population. Accordingly, we observed lower levels of phosphorylated RB (Ser807/811) and an increased BAX/BCL2 ratio in RNAi treated MCF7 cells. We also showed that CHRNA5 RNAi transcriptome correlated negatively with DDR relevant gene expression profile in breast cancer gene expression datasets while the coexposure to topoisomerase inhibitors in the presence of CHRNA5 RNAi enhanced chemosensitivity potentially due to reduced DDR. CHRNA5 RNAi consistently lowered total CHEK1 mRNA and protein levels as well as phosphorylated CHEK1 (Ser345) in MCF7 cells. We also detected a significant positive correlation between the expression levels of CHRNA5 and CHEK1 in CCLE, TCGA and METABRIC breast cancer datasets. Our study suggests CHRNA5 RNAi is associated with cell cycle inhibition, apoptosis as well as reduced DDR and increased drug sensitivity in breast cancer yet future studies are warranted since dose- and cell line-specific differences exist in response to CHRNA5 depletion. Gene expression microarray data can be accessed from GEO database under the accession number GSE89333.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/pathology , Cell Cycle/genetics , DNA Damage/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , RNA Interference , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Signal Transduction/genetics
11.
Genet Test Mol Biomarkers ; 22(12): 714-718, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30461311

ABSTRACT

Background: Escobar syndrome, a nonlethal variant of multiple pterygium syndromes (MPS), is a rare autosomal recessive disorder characterized by pterygia and multiple joint contractures along with other anomalies. Variants in cholinergic receptor nicotinic gamma subunit (CHRNG) have been previously reported in patients with Escobar syndrome. Objective: We studied a consanguineous Pakistani family affected with Escobar syndrome to identify the underlying genetic defect through short tandem repeat (STR) genotyping and direct DNA sequencing. Results: Genotyping with microsatellite markers (D2S427, D2S2344, and D2S206) revealed linkage of the disease phenotype in the family to the CHRNG locus. Using Sanger sequencing, we identified a homozygous nonsense CHRNG variant c.136C>T (p.R46*), predicted to produce a truncated protein that leads to acetylcholine receptor deficiency, causing MPS. The unaffected parents and siblings in the family were heterozygous carriers of this disease-causing variant. Conclusion: We report the identification of a nonsense CHRNG variant in a consanguineous Pakistani family affected with Escobar syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Codon, Nonsense , Malignant Hyperthermia/genetics , Receptors, Nicotinic/genetics , Skin Abnormalities/genetics , Abnormalities, Multiple/ethnology , Consanguinity , Female , Genes, Recessive , Genotype , Humans , Male , Malignant Hyperthermia/ethnology , Microsatellite Repeats , Pakistan , Pedigree , Phenotype , Receptors, Nicotinic/deficiency , Skin Abnormalities/ethnology
12.
Basic Res Cardiol ; 113(5): 38, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30097758

ABSTRACT

The CHRNA5 gene encodes a neurotransmitter receptor subunit involved in multiple processes, including cholinergic autonomic nerve activity and inflammation. Common variants in CHRNA5 have been linked with atherosclerotic cardiovascular disease. Association of variation in CHRNA5 and specific haplotypes with cardiovascular outcomes has not been described. The aim of this study was to examine the association of CHRNA5 haplotypes with gene expression and mortality among patients with acute myocardial infarction (AMI) and explore potential mechanisms of this association. Patients (N = 2054) hospitalized with AMI were genotyped for two common variants in CHRNA5. Proportional hazard models were used to estimate independent association of CHRNA5 haplotype with 1-year mortality. Both individual variants were associated with mortality (p = 0.0096 and 0.0004, respectively) and were in tight LD (D' = 0.99). One haplotype, HAP3, was associated with decreased mortality one year after AMI (adjusted HR = 0.42, 95% CI 0.26, 0.68; p = 0.0004). This association was validated in an independent cohort (N = 637) of post-MI patients (adjusted HR = 0.23, 95% CI 0.07, 0.79; p = 0.019). Differences in CHRNA5 expression by haplotype were investigated in human heart samples (n = 28). Compared with non-carriers, HAP3 carriers had threefold lower cardiac CHRNA5 mRNA expression (p = 0.023). Circulating levels of the inflammatory marker hsCRP were significantly lower in HAP3 carriers versus non-carriers (3.43 ± 4.2 versus 3.91 ± 5.1; p = 0.0379). Activation of the inflammasome, an important inflammatory complex involved in cardiovascular disease that is necessary for release of the pro-inflammatory cytokine IL-1 ß, was assessed in bone marrow-derived macrophages (BMDM) from CHRNA5 knockout mice and wild-type controls. In BMDM from CHRNA5 knockout mice, IL-1ß secretion was reduced by 50% compared to wild-type controls (p = 0.004). Therefore, a common haplotype of CHRNA5 that results in reduced cardiac expression of CHRNA5 and attenuated macrophage inflammasome activation is associated with lower mortality after AMI. These results implicate CHRNA5 and the cholinergic anti-inflammatory pathway in survival following AMI.


Subject(s)
Myocardial Infarction/genetics , Myocarditis/genetics , Nerve Tissue Proteins/genetics , Receptors, Nicotinic/genetics , Aged , Animals , Cells, Cultured , Female , Genetic Association Studies , Genetic Markers , Genetic Predisposition to Disease , Haplotypes , Humans , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocardial Infarction/diagnosis , Myocardial Infarction/metabolism , Myocardial Infarction/mortality , Myocarditis/diagnosis , Myocarditis/metabolism , Myocarditis/mortality , Phenotype , Prognosis , Prospective Studies , Protective Factors , Receptors, Nicotinic/deficiency , Risk Factors , Time Factors , United States/epidemiology
13.
J Neurosci ; 38(34): 7440-7451, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30030403

ABSTRACT

Cochlear synaptopathy produced by exposure to noise levels that cause only transient auditory threshold elevations is a condition that affects many people and is believed to contribute to poor speech discrimination in noisy environments. These functional deficits in hearing, without changes in sensitivity, have been called hidden hearing loss (HHL). It has been proposed that activity of the medial olivocochlear (MOC) system can ameliorate acoustic trauma effects. Here we explore the role of the MOC system in HHL by comparing the performance of two different mouse models: an α9 nicotinic receptor subunit knock-out (KO; Chrna9 KO), which lacks cholinergic transmission between efferent neurons and hair cells; and a gain-of-function knock-in (KI; Chrna9L9'T KI) carrying an α9 point mutation that leads to enhanced cholinergic activity. Animals of either sex were exposed to sound pressure levels that in wild-type produced transient cochlear threshold shifts and a decrease in neural response amplitudes, together with the loss of ribbon synapses, which is indicative of cochlear synaptopathy. Moreover, a reduction in the number of efferent contacts to outer hair cells was observed. In Chrna9 KO ears, noise exposure produced permanent auditory threshold elevations together with cochlear synaptopathy. In contrast, the Chrna9L9'T KI was completely resistant to the same acoustic exposure protocol. These results show a positive correlation between the degree of HHL prevention and the level of cholinergic activity. Notably, enhancement of the MOC feedback promoted new afferent synapse formation, suggesting that it can trigger cellular and molecular mechanisms to protect and/or repair the inner ear sensory epithelium.SIGNIFICANCE STATEMENT Noise overexposure is a major cause of a variety of perceptual disabilities, including speech-in-noise difficulties, tinnitus, and hyperacusis. Here we show that exposure to noise levels that do not cause permanent threshold elevations or hair cell death can produce a loss of cochlear nerve synapses to inner hair cells as well as degeneration of medial olivocochlear (MOC) terminals contacting the outer hair cells. Enhancement of the MOC reflex can prevent both types of neuropathy, highlighting the potential use of drugs that increase α9α10 nicotinic cholinergic receptor activity as a pharmacotherapeutic strategy to avoid hidden hearing loss.


Subject(s)
Auditory Threshold/physiology , Cochlea/physiopathology , Hearing Loss, Noise-Induced/physiopathology , Olivary Nucleus/physiopathology , Receptors, Nicotinic/physiology , Animals , Auditory Pathways/physiopathology , Cholinergic Fibers/physiology , Efferent Pathways/physiopathology , Feedback, Physiological , Gain of Function Mutation , Hair Cells, Auditory, Outer/physiology , Hearing Loss, Noise-Induced/etiology , Humans , Mice , Nerve Regeneration , Noise/adverse effects , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Synapses/physiology
14.
Neuropharmacology ; 138: 371-380, 2018 08.
Article in English | MEDLINE | ID: mdl-29940207

ABSTRACT

Preclinical studies suggest the involvement of various subtypes of nicotinic acetylcholine receptors in the pathophysiology of Parkinson's disease, a neurodegenerative disorder characterized by the death of dopaminergic neurons in the substantia nigra pars compacta (SNC). We studied for the first time the effects of α5 nicotinic receptor subunit gene deletion on motor behavior and neurodegeneration in mouse models of Parkinson's disease and levodopa-induced dyskinesia. Unilateral dopaminergic lesions were induced in wild-type and α5-KO mice by 6-hydroxydopamine injections into the striatum or the medial forebrain bundle. Subsequently, rotational behavior induced by dopaminergic drugs was measured. A subset of animals received chronic treatments with levodopa and nicotine to assess levodopa-induced dyskinesia and antidyskinetic effects by nicotine. SNC lesion extent was assessed with tyrosine hydroxylase immunohistochemistry and stereological cell counting. Effects of α5 gene deletion on the dopaminergic system were investigated by measuring ex vivo striatal dopamine transporter function and protein expression, dopamine and metabolite tissue concentrations and dopamine receptor mRNA expression. Hemiparkinsonian α5-KO mice exhibited attenuated rotational behavior after amphetamine injection and attenuated levodopa-induced dyskinesia. In the intrastriatal lesion model, dopaminergic cell loss in the medial cluster of the SNC was less severe in α5-KO mice. Decreased striatal dopamine uptake in α5-KO animals suggested reduced dopamine transporter function as a mechanism of attenuated neurotoxicity. Nicotine reduced dyskinesia severity in wild-type but not α5-KO mice. The attenuated dopaminergic neurodegeneration and motor dysfunction observed in hemiparkinsonian α5-KO mice suggests potential for α5 subunit-containing nicotinic receptors as a novel target in the treatment of Parkinson's disease.


Subject(s)
Dopaminergic Neurons/metabolism , Motor Activity/physiology , Neurodegenerative Diseases/metabolism , Parkinsonian Disorders/metabolism , Receptors, Nicotinic/deficiency , Amphetamine/pharmacology , Animals , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Brain/drug effects , Brain/metabolism , Brain/pathology , Central Nervous System Stimulants/pharmacology , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dyskinesia, Drug-Induced/metabolism , Female , Functional Laterality , Levodopa/adverse effects , Levodopa/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Neurodegenerative Diseases/pathology , Oxidopamine , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , RNA, Messenger/metabolism , Receptors, Dopamine/metabolism , Receptors, Nicotinic/genetics
15.
Neuropharmacology ; 138: 341-348, 2018 08.
Article in English | MEDLINE | ID: mdl-29944862

ABSTRACT

Evidence suggests that there is an association between polymorphisms in the α5 nicotinic acetylcholine receptor (nAChR) subunit and risk of developing alcohol dependence in humans. The α5 nAChR subunit has also recently been shown to modulate some of the acute response to ethanol in mice. The aim of the current study was to further characterize the role of α5-containing (α5*) nAChRs in acute ethanol responsive behaviors, ethanol consumption and ethanol preference in mice. We conducted a battery of tests in male α5 knockout (KO) mice for a range of ethanol-induced behaviors including hypothermia, hypnosis, and anxiolysis. We also investigated the effects of α5* nAChR on ethanol reward using the Conditioned Place Preference (CPP) assay. Further, we tested the effects of gene deletion on drinking behaviors using the voluntary ethanol consumption in a two-bottle choice assay and Drinking in the Dark (DID, with or without stress) paradigm. We found that deletion of the α5 nAChR subunit enhanced ethanol-induced hypothermia, hypnosis, and an anxiolytic-like response in comparison to wild-type controls. The α5 KO mice showed reduced CPP for ethanol, suggesting that the rewarding properties of ethanol are decreased in mutant mice. Interestingly, Chrna5 gene deletion had no effect on basal ethanol drinking behavior, or ethanol metabolism, but did decrease ethanol intake in the DID paradigm following restraint stress. Taken together, we provide new evidence that α5 nAChRs are involved in some but not all of the behavioral effects of ethanol. Our results highlight the importance of nAChRs as a possible target for the treatment of alcohol dependence.


Subject(s)
Alcohol Drinking/metabolism , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Receptors, Nicotinic/deficiency , Reward , Alcohol Drinking/psychology , Animals , Anxiety/chemically induced , Anxiety/metabolism , Central Nervous System Depressants/blood , Choice Behavior/drug effects , Choice Behavior/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Ethanol/blood , Hypnotics and Sedatives/blood , Hypnotics and Sedatives/pharmacology , Hypothermia/chemically induced , Hypothermia/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Receptors, Nicotinic/genetics , Reflex/drug effects , Reflex/physiology , Spatial Behavior/drug effects , Spatial Behavior/physiology
16.
J Neurosci ; 38(31): 6900-6920, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29954848

ABSTRACT

Genetic studies have shown an association between smoking and variation at the CHRNA5/A3/B4 gene locus encoding the α5, α3, and ß4 nicotinic receptor subunits. The α5 receptor has been specifically implicated because smoking-associated haplotypes contain a coding variant in the CHRNA5 gene. The Chrna5/a3/b4 locus is conserved in rodents and the restricted expression of these subunits suggests neural pathways through which the reinforcing and aversive properties of nicotine may be mediated. Here, we show that, in the interpeduncular nucleus (IP), the site of the highest Chrna5 mRNA expression in rodents, electrophysiological responses to nicotinic acetylcholine receptor stimulation are markedly reduced in α5-null mice. IP neurons differ markedly from their upstream ventral medial habenula cholinergic partners, which appear unaltered by loss of α5. To probe the functional role of α5-containing IP neurons, we used BAC recombineering to generate transgenic mice expressing Cre-recombinase from the Chrna5 locus. Reporter expression driven by Chrna5Cre demonstrates that transcription of Chrna5 is regulated independently from the Chrna3/b4 genes transcribed on the opposite strand. Chrna5-expressing IP neurons are GABAergic and project to distant targets in the mesopontine raphe and tegmentum rather than forming local circuits. Optogenetic stimulation of Chrna5-expressing IP neurons failed to elicit physical manifestations of withdrawal. However, after recent prior stimulation or exposure to nicotine, IP stimulation becomes aversive. These results using mice of both sexes support the idea that the risk allele of CHRNA5 may increase the drive to smoke via loss of IP-mediated nicotine aversion.SIGNIFICANCE STATEMENT Understanding the receptors and neural pathways underlying the reinforcing and aversive effects of nicotine may suggest new treatments for tobacco addiction. Part of the individual variability in smoking is associated with specific forms of the α5 nicotinic receptor subunit gene. Here, we show that deletion of the α5 subunit in mice markedly reduces the cellular response to nicotine and acetylcholine in the interpeduncular nucleus (IP). Stimulation of α5-expressing IP neurons using optogenetics is aversive, but this effect requires priming by recent prior stimulation or exposure to nicotine. These results support the idea that the smoking-associated variant of the α5 gene may increase the drive to smoke via loss of IP-mediated nicotine aversion.


Subject(s)
Avoidance Learning/physiology , Interpeduncular Nucleus/physiology , Nicotine/pharmacology , Receptors, Nicotinic/physiology , Smoking/psychology , Animals , Crosses, Genetic , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Genes, Reporter , Injections, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nicotine/administration & dosage , Nicotine/toxicity , Optogenetics , Patch-Clamp Techniques , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Recombinant Fusion Proteins/metabolism , Smoking/genetics , Smoking/physiopathology , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/physiopathology
17.
Neuropharmacology ; 135: 529-535, 2018 06.
Article in English | MEDLINE | ID: mdl-29677582

ABSTRACT

The objective of the current study is to test the hypothesis that the deletion of alpha(α)2* nicotinic acetylcholine receptors (nAChRs) (encoded by the Chrna2 gene) ablate maternal nicotine-induced learning and memory deficits in adolescent mice. We use a pre-exposure-dependent contextual fear conditioning behavioral paradigm that is highly hippocampus-dependent. Adolescent wild type and α2-null mutant offspring are exposed to vehicle or maternal nicotine exposure (200 µg/ml, expressed as base) in the drinking water throughout pregnancy until weaning. Adolescent male offspring mice are tested for alterations in growth and development characteristics as well as modifications in locomotion, anxiety, shock-reactivity and learning and memory. As expected, maternal nicotine exposure has no effects on pup number, weight gain and only modestly reduces fluid intake by 19%. Behaviorally, maternal nicotine exposure impedes extinction learning in adolescent wild type mice, a consequence that is abolished in α2-null mutant mice. The effects on learning and memory are not confounded by alternations in stereotypy, locomotion, anxiety or sensory shock reactivity. Overall, the findings highlight that the deletion of α2* nAChRs eliminate the effects of maternal nicotine exposure on learning and memory in adolescent mice.


Subject(s)
Learning/physiology , Maternal Exposure , Memory/physiology , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Receptors, Nicotinic/deficiency , Animals , Anxiety/metabolism , Female , Learning/drug effects , Male , Memory/drug effects , Memory Disorders/etiology , Memory Disorders/metabolism , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Receptors, Nicotinic/genetics
18.
Br J Pharmacol ; 175(11): 1944-1956, 2018 06.
Article in English | MEDLINE | ID: mdl-28585241

ABSTRACT

BACKGROUND AND PURPOSE: Genomic analysis has shown many variants in both CHRNA4 and CHRNB2, genes which encode the α4 and ß2 subunits of nicotinic ACh receptors (nAChR) respectively. Some variants influence receptor expression, raising the possibility that CHRNA4 variants may affect response to tobacco use in humans. Chronic exposure to nicotine increases expression of nAChRs, particularly α4ß2-nAChRs, in humans and laboratory animals. Here, we have evaluated whether the initial level of receptor expression affects the increase in expression. EXPERIMENTAL APPROACH: Mice differing in expression of α4 and/or ß2 nAChR subunits were chronically treated with saline, 0.25, 1.0 or 4.0 mg·kg-1 ·h-1 nicotine. Brain preparations were analysed autoradiographically by [125 I]-epibatidine binding, immunoprecipitation and Western blotting. KEY RESULTS: Immunochemical studies confirmed that most of the [3 H]-epibatidine binding corresponds to α4ß2*-nAChR and that increases in binding correspond to increases in α4 and ß2 proteins. Consistent with previous reports, the dose-dependent increase in nAChR in wild-type mice following chronic nicotine treatment, measured with any of the methods, reached a maximum. Although receptor expression was reduced by approximately 50% in ß2+- mice, the pattern of response to chronic treatment resembled that of wild-type mice. In contrast, both α4+- and α4+- /ß2+- exhibited relatively greater up-regulation. Consistent with previous reports, α4ß2α5-nAChR did not increase in response to nicotine. CONCLUSIONS AND IMPLICATIONS: These results indicate that mice with reduced expression of the α4 nAChR subunit have a more robust response to chronic nicotine than mice with normal expression of this subunit. LINKED ARTICLES: This article is part of a themed section on Nicotinic Acetylcholine Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.11/issuetoc.


Subject(s)
Nicotine/pharmacology , Receptors, Nicotinic/metabolism , Up-Regulation/drug effects , Animals , Dose-Response Relationship, Drug , Female , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotine/administration & dosage , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Structure-Activity Relationship
19.
Behav Brain Res ; 328: 105-114, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28408300

ABSTRACT

The α9α10-subtype of nicotinic acetylcholine receptor (nAChR) has recently garnered interest in biomedicine and is being pursued as an analgesic target. However, the receptor exhibits diverse tissue distribution, the function of which is known to varying degrees, and targeting this receptor for clinical treatments without a broad understanding of its function may have adverse consequences. The α9α10-nAChR is expressed in the adrenal and pituitary glands, suggesting a potential role in the stress response, but little is known about its function in this tissue. Here we determined a role for the α9α10-nAChR in behavioural and physiological stress responses, by comparing the stress- and affect-related phenotypes of wildtype and α9-nAChR knockout mice. Naïve knockout mice exhibited largely normal behaviour on standard tests of affective behaviour. However, after sub-chronic restraint stress knockout mice showed significantly decreased stress-induced arousal and increased anxiety-like behaviour when compared to wildtype animals. Physiologically, corticosterone responses were muted in knockout mice after an acute stressor, but exaggerated in response to the same stressor after undergoing sub-chronic stress. Behavioural profiling of the α9-nAChR knockout mice in the home-cage revealed that circadian patterns of activity were altered when compared to wildtype controls. Furthermore, knockout mice showed altered responses to a period of reward discounting, resulting in anhedonia-like behaviour in a sucrose preference test where WT mice continued to seek reward. These experiments uncover a novel role for the α9α10-nAChR in mounting a normal stress response and in the regulation of affective- and reward-related behaviour, and suggest that pursuing the receptor for clinical treatments may not be as straightforward as has been suggested.


Subject(s)
Affect/physiology , Receptors, Nicotinic/deficiency , Reward , Stress, Psychological/metabolism , Anhedonia/physiology , Animals , Anxiety/metabolism , Arousal/physiology , Circadian Rhythm/physiology , Corticosterone/blood , Male , Mice, 129 Strain , Mice, Knockout , Receptors, Nicotinic/genetics
20.
J Neurophysiol ; 117(4): 1553-1568, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28077670

ABSTRACT

The α9-nicotinic acetylcholine receptor (α9-nAChR) subunit is expressed in the vestibular and auditory periphery, and its loss of function could compromise peripheral input from the predominantly cholinergic efferent vestibular system (EVS). A recent study has shown that α9-nAChRs play an important role in short-term vestibulo-ocular reflex (VOR) adaptation. We hypothesize that α9-nAChRs could also be important for other forms of vestibular plasticity, such as that needed for VOR recovery after vestibular organ injury. We measured the efficacy of VOR compensation in α9 knockout mice. These mice have deletion of most of the gene (chrna9) encoding the nAChR and thereby lack α9-nAChRs. We measured the VOR gain (eye velocity/head velocity) in 20 α9 knockout mice and 16 cba129 controls. We measured the sinusoidal (0.2-10 Hz, 20-100°/s) and transient (1,500-6,000°/s2) VOR in complete darkness before (baseline) unilateral labyrinthectomy (UL) and then 1, 5, and 28 days after UL. On day 1 after UL, cba129 mice retained ~50% of their initial function for contralesional rotations, whereas α9 knockout mice only retained ~20%. After 28 days, α9 knockout mice had ~50% lower gain for both ipsilesional and contralesional rotations compared with cba129 mice. Cba129 mice regained ~75% of their baseline function for ipsilesional and ~90% for contralesional rotations. In contrast, α9 knockout mice only regained ~30% and ~50% function, respectively, leaving the VOR severely impaired for rotations in both directions. Our results show that loss of α9-nAChRs severely affects VOR compensation, suggesting that complimentary central and peripheral EVS-mediated adaptive mechanisms might be affected by this loss.NEW & NOTEWORTHY Loss of the α9-nicotinic acetylcholine receptor (α9-nAChR) subunit utilized by the efferent vestibular system (EVS) has been shown to significantly affect vestibulo-ocular reflex (VOR) adaptation. In our present study we have shown that loss of α9-nAChRs also affects VOR compensation, suggesting that the mammalian EVS plays an important role in vestibular plasticity, in general, and that VOR compensation is a more distributed process than previously thought, relying on both central and peripheral changes.


Subject(s)
Adaptation, Physiological/physiology , Efferent Pathways/physiology , Functional Laterality/physiology , Reflex, Vestibulo-Ocular/physiology , Vestibule, Labyrinth/physiology , Adaptation, Physiological/genetics , Animals , Female , Functional Laterality/genetics , Linear Models , Male , Mice , Mice, Inbred CBA , Mice, Knockout , Nystagmus, Physiologic , Reaction Time , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Rotation , Time Factors , Vestibule, Labyrinth/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...