Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Glia ; 72(6): 1183-1200, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38477581

ABSTRACT

Barrier-forming olfactory glia cells, termed sustentacular cells, play important roles for immune defense of the olfactory mucosa, for example as entry sites for SARS-CoV-2 and subsequent development of inflammation-induced smell loss. Here we demonstrate that sustentacular cells express ACKR3, a chemokine receptor that functions both as a scavenger of the chemokine CXCL12 and as an activator of alternative signaling pathways. Differential gene expression analysis of bulk RNA sequencing data obtained from WT and ACKR3 conditional knockout mice revealed upregulation of genes involved in immune defense. To map the regulated genes to the different cell types of the olfactory mucosa, we employed biocomputational methods utilizing a single-cell reference atlas. Transcriptome analysis, PCR and immunofluorescence identified up-regulation of NF-κB-related genes, known to amplify inflammatory signaling and to facilitate leukocyte transmigration, in the gliogenic lineage. Accordingly, we found a marked increase in leukocyte-expressed genes and confirmed leukocyte infiltration into the olfactory mucosa. In addition, lack of ACKR3 led to enhanced expression and secretion of early mediators of immune defense by Bowman's glands. As a result, the number of apoptotic cells in the epithelium was decreased. In conclusion, our research underlines the importance of sustentacular cells in immune defense of the olfactory mucosa. Moreover, it identifies ACKR3, a druggable G protein-coupled receptor, as a promising target for modulation of inflammation-associated anosmia.


Subject(s)
Inflammation , Olfactory Mucosa , Animals , Mice , Chemokine CXCL12/metabolism , Gene Expression Profiling , Inflammation/metabolism , Neuroglia/metabolism , Olfactory Mucosa/metabolism
2.
Stem Cells ; 39(5): 617-635, 2021 05.
Article in English | MEDLINE | ID: mdl-33470495

ABSTRACT

The olfactory epithelium (OE) possesses unique lifelong neuroregenerative capacities and undergoes constitutive neurogenesis throughout mammalian lifespan. Two populations of stem cells, frequently dividing globose basal cells (GBCs) and quiescent horizontal basal cells (HBCs), readily replace olfactory neurons throughout lifetime. Although lineage commitment and neuronal differentiation of stem cells has already been described in terms of transcription factor expression, little is known about external factors balancing between differentiation and self-renewal. We show here that expression of the CXC-motif chemokine receptor 4 (CXCR4) distinguishes both types of stem cells. Extensive colocalization analysis revealed exclusive expression of CXCR4 in proliferating GBCs and their neuronal progenies. Moreover, only neuronal lineage cells were derived from CXCR4-CreER-tdTomato reporter mice in the OE. Furthermore, Cre-tdTomato mice specific for HBCs (Nestin+ and Cytokeratin14+) did not reduce CXCR4 expression when bred to mice bearing floxed CXCR4 alleles, and did not show labeling of the neuronal cells. CXCR4 and its ligand CXCL12 were markedly upregulated upon induction of GBC proliferation during injury-induced regeneration. in vivo overexpression of CXCL12 did downregulate CXCR4 levels, which results in reduced GBC maintenance and neuronal differentiation. We proved that these effects were caused by CXCR4 downregulation rather than over-activation by showing that the phenotypes of CXCL12-overexpressing mice were highly similar to the phenotypes of CXCR4 knockout mice. Our results demonstrate functional CXCR4 signaling in GBCs regulates cell cycle exit and neural differentiation. We propose that CXCR4/CXCL12 signaling is an essential regulator of olfactory neurogenesis and provide new insights into the dynamics of neurogenesis in the OE.


Subject(s)
Chemokine CXCL12/genetics , Nerve Regeneration/genetics , Neurogenesis/genetics , Olfactory Nerve/growth & development , Receptors, CXCR4/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Keratin-14/genetics , Mice , Mice, Knockout , Nestin/genetics , Neural Stem Cells/cytology , Neurons/cytology , Olfactory Mucosa/growth & development , Olfactory Mucosa/injuries , Olfactory Nerve/metabolism
3.
PLoS One ; 13(2): e0191219, 2018.
Article in English | MEDLINE | ID: mdl-29444117

ABSTRACT

TMEM16 proteins are a recently identified protein family comprising Ca2+-activated Cl- channels that generate outwardly rectifying ionic currents in response to intracellular Ca2+ elevations. Some TMEM16 family members, such as TMEM16F/ANO6 are also essential for Ca2+-dependent phospholipid scrambling. TMEM16-like genes are present in the genomes of most eukaryotic species, the function(s) of TMEM16 family members from evolutionary ancient eukaryotes is not completely clear. Here, we provide insight into the evolution of these TMEM16 proteins by similarity searches for ancestral sequences. All eukaryotic genomes contain TMEM16 homologs, but only vertebrates have the full repertoire of ten distinct subtypes. TMEM16 homologs studied so far belong to the opisthokont branch of the phylogenetic tree, which includes the animal and fungal kingdoms. An organism outside this group is Dictyostelium discoideum, a representative of the amoebozoa group that diverged from the metazoa before fungi. We here functionally investigated the TMEM16 family member from Dictyostelium discoideum. When recombinantly expressed in HEK293 cells, DdTMEM16 induces phospholipid scrambling. However, in several electrophysiological experiments we did not find evidence for a Ca2+-activated Cl- channel function of DdTMEM16.


Subject(s)
Anoctamins/genetics , Dictyostelium/genetics , Animals , Anoctamins/metabolism , Biological Evolution , Chloride Channels/genetics , Chloride Channels/metabolism , Dictyostelium/metabolism , Eukaryota/genetics , Evolution, Molecular , HEK293 Cells , Humans , Ion Transport/genetics , Ion Transport/physiology , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism , Phospholipids/physiology , Phylogeny , Recombinant Proteins
4.
Sci Rep ; 7(1): 8409, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814779

ABSTRACT

BEACH domain proteins are involved in membrane protein traffic and human diseases, but their molecular mechanisms are not understood. The BEACH protein LRBA has been implicated in immune response and cell proliferation, and human LRBA mutations cause severe immune deficiency. Here, we report a first functional and molecular phenotype outside the immune system of LRBA-knockout mice: compromised olfaction, manifesting in reduced electro-olfactogram response amplitude, impaired food-finding efficiency, and smaller olfactory bulbs. LRBA is prominently expressed in olfactory and vomeronasal chemosensory neurons of wild-type mice. Olfactory impairment in the LRBA-KO is explained by markedly reduced concentrations (20-40% of wild-type levels) of all three subunits αolf, ß1 and γ13 of the olfactory heterotrimeric G-protein, Golf, in the sensory cilia of olfactory neurons. In contrast, cilia morphology and the concentrations of many other proteins of olfactory cilia are not or only slightly affected. LRBA is also highly expressed in photoreceptor cells, another cell type with a specialized sensory cilium and heterotrimeric G-protein-based signalling; however, visual function appeared unimpaired by the LRBA-KO. To our knowledge, this is the first observation that a BEACH protein is required for the efficient subcellular localization of a lipid-anchored protein, and of a ciliary protein.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cilia/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Electroretinography , Female , Gene Expression Regulation , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Male , Mice, Knockout , Mice, Transgenic , Olfaction Disorders/genetics , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Olfactory Receptor Neurons/metabolism , Protein Domains , Retina/abnormalities , Vomeronasal Organ/cytology , Vomeronasal Organ/metabolism
5.
Exp Dermatol ; 26(7): 569-576, 2017 07.
Article in English | MEDLINE | ID: mdl-28191688

ABSTRACT

We identified the olfactory receptor 51E2 in human melanoma and have measured both OR51E2 mRNA and protein expression in melanoma tissue sections. qPCR analysis revealed that the receptor is upregulated in melanoma cells compared to normal melanocytes, indicating that OR51E2 may play a role in early melanoma development and progression. Activation of endogenous OR51E2 in cultured cells derived from metastatic and vertical-growth phase (VGP) by its ligand ß-ionone results in an increase in the intracellular Ca2+ concentration. RNAi experiments showed that the ß-ionone-induced Ca2+ signal depends on the activation of OR51E2. Furthermore, OR51E2 activation inhibits the growth of VGP melanoma cells via apoptotic processes. Cell motility assays revealed that treatment with ß-ionone decreases the migration of VGP melanoma cells. Overall, our data demonstrates that OR51E2 is involved in the regulation of cell proliferation and migration, suggesting that it may serve as a novel target for melanoma therapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Neoplasm Proteins/metabolism , Receptors, Odorant/metabolism , Skin Neoplasms/metabolism , Calcium/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Humans , Ligands , Melanocytes/metabolism , Neoplasm Metastasis , Norisoprenoids/chemistry , Polymerase Chain Reaction , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction , Treatment Outcome
6.
Chem Senses ; 42(1): 25-35, 2017 01.
Article in English | MEDLINE | ID: mdl-27655939

ABSTRACT

In most mammals, the vomeronasal system detects a variety of (semio)chemicals that mediate olfactory-driven social and sexual behaviors. Vomeronasal chemosensation depends on G protein-coupled receptors (V1R, V2R, and FPR-rs) that operate at remarkably low stimulus concentrations, thus, indicating a highly sensitive and efficient signaling pathway. We identified the PDZ domain-containing protein, Na+/H+ exchanger regulatory factor-1 (NHERF1), as putative molecular organizer of signal transduction in vomeronasal neurons. NHERF1 is a protein that contains 2 PDZ domains and a carboxy-terminal ezrin-binding domain. It localizes to microvilli of vomeronasal sensory neurons and interacts with V1Rs. Furthermore, NHERF1 and Gαi2 are closely colocalized. These findings open up new aspects of the functional organization and regulation of vomeronasal signal transduction by PDZ scaffolding proteins.


Subject(s)
Microvilli/chemistry , Phosphoproteins/analysis , Sensory Receptor Cells/chemistry , Sodium-Hydrogen Exchangers/analysis , Vomeronasal Organ/cytology , Animals , Mice , Mice, Inbred C57BL , Microvilli/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Sensory Receptor Cells/metabolism , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
7.
Chem Senses ; 41(8): 669-76, 2016 10.
Article in English | MEDLINE | ID: mdl-27377750

ABSTRACT

In rodents, the vomeronasal system controls social and sexual behavior. However, several mechanistic aspects of sensory signaling in the vomeronasal organ remain unclear. Here, we investigate the biophysical basis of a recently proposed vomeronasal signal transduction component-a Ca(2+)-activated Cl(-) current. As the physiological role of such a current is a direct function of the Cl(-) equilibrium potential, we determined the intracellular Cl(-) concentration in dendritic knobs of vomeronasal neurons. Quantitative fluorescence lifetime imaging of a Cl(-)-sensitive dye at the apical surface of the intact vomeronasal neuroepithelium revealed increased cytosolic Cl(-) levels in dendritic knobs, a substantially lower Cl(-) concentration in vomeronasal sustentacular cells, and an apparent Cl(-) gradient in vomeronasal neurons along their dendritic apicobasal axis. Together, our data provide a biophysical basis for sensory signal amplification in vomeronasal neuron microvilli by opening Ca(2+)-activated Cl(-) channels.


Subject(s)
Chlorides/analysis , Cytosol/chemistry , Dendrites/chemistry , Sensory Receptor Cells/chemistry , Vomeronasal Organ/chemistry , Animals , Calcium/metabolism , Chloride Channels/metabolism , Cytosol/metabolism , Dendrites/metabolism , Mice , Sensory Receptor Cells/metabolism , Vomeronasal Organ/metabolism
8.
J Biol Chem ; 291(34): 17772-86, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27226631

ABSTRACT

Olfactory receptors, which belong to the family of G-protein-coupled receptors, are found to be ectopically expressed in non-sensory tissues mediating a variety of cellular functions. In this study we detected the olfactory receptor OR51E2 at the transcript and the protein level in human epidermal melanocytes. Stimulation of primary melanocytes with the OR51E2 ligand ß-ionone significantly inhibited melanocyte proliferation. Our results further showed that ß-ionone stimulates melanogenesis and dendritogenesis. Using RNA silencing and receptor antagonists, we demonstrated that OR51E2 activation elevated cytosolic Ca(2+) and cAMP, which could mediate the observed increase in melanin synthesis. Co-immunocytochemical stainings using a specific OR51E2 antibody revealed subcellular localization of the receptor in early endosomes associated with EEA-1 (early endosome antigen 1). Plasma membrane preparations showed that OR51E2 protein is present at the melanocyte cell surface. Our findings thus suggest that activation of olfactory receptor signaling by external compounds can influence melanocyte homeostasis.


Subject(s)
Calcium Signaling/physiology , Cell Membrane/metabolism , Epidermis/metabolism , Melanocytes/metabolism , Neoplasm Proteins/metabolism , Receptors, Odorant/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Cell Line , Cyclic AMP/metabolism , Endosomes/metabolism , Epidermal Cells , Humans , Melanocytes/cytology , Norisoprenoids/pharmacology , Vesicular Transport Proteins/metabolism
9.
Front Cell Neurosci ; 9: 366, 2015.
Article in English | MEDLINE | ID: mdl-26441537

ABSTRACT

Olfactory signals influence food intake in a variety of species. To maximize the chances of finding a source of calories, an animal's preference for fatty foods and triglycerides already becomes apparent during olfactory food search behavior. However, the molecular identity of both receptors and ligands mediating olfactory-dependent fatty acid recognition are, so far, undescribed. We here describe that a subset of olfactory sensory neurons expresses the fatty acid receptor CD36 and demonstrate a receptor-like localization of CD36 in olfactory cilia by STED microscopy. CD36-positive olfactory neurons share olfaction-specific transduction elements and project to numerous glomeruli in the ventral olfactory bulb. In accordance with the described roles of CD36 as fatty acid receptor or co-receptor in other sensory systems, the number of olfactory neurons responding to oleic acid, a major milk component, in Ca(2+) imaging experiments is drastically reduced in young CD36 knock-out mice. Strikingly, we also observe marked age-dependent changes in CD36 localization, which is prominently present in the ciliary compartment only during the suckling period. Our results support the involvement of CD36 in fatty acid detection by the mammalian olfactory system.

10.
J Biol Rhythms ; 30(6): 506-18, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26482709

ABSTRACT

Odor discrimination behavior displays circadian fluctuations in mice, indicating that mammalian olfactory function is under control of the circadian system. This is further supported by the facts that odor discrimination rhythms depend on the presence of clock genes and that olfactory tissues contain autonomous circadian clocks. However, the molecular link between circadian function and olfactory processing is still unknown. To elucidate the molecular mechanisms underlying this link, we focused on the olfactory epithelium (OE), the primary target of odors and the site of the initial events in olfactory processing. We asked whether olfactory sensory neurons (OSNs) within the OE possess an autonomous circadian clock and whether olfactory pathways are under circadian control. Employing clock gene-driven bioluminescence reporter assays and time-dependent immunohistochemistry on OE samples, we found robust circadian rhythms of core clock genes and their proteins in OSNs, suggesting that the OE indeed contains an autonomous circadian clock. Furthermore, we performed a circadian transcriptome analysis and identified several OSN-specific components that are under circadian control, including those with putative roles in circadian olfactory processing, such as KIRREL2-an established factor involved in short-term OSN activation. The spatiotemporal expression patterns of our candidate proteins suggest that they are involved in short-term anabolic processes to rhythmically prepare the cell for peak performances and to promote circadian function of OSNs.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/genetics , Odorants , Olfactory Mucosa/physiology , Olfactory Receptor Neurons/physiology , Animals , Circadian Clocks/physiology , Circadian Rhythm/radiation effects , Gene Expression Regulation , Immunoglobulins/genetics , Luminescent Measurements , Membrane Proteins/genetics , Mice , Olfactory Pathways/physiology , Period Circadian Proteins/genetics , Time Factors , Tissue Array Analysis
11.
PLoS One ; 10(1): e0113170, 2015.
Article in English | MEDLINE | ID: mdl-25590618

ABSTRACT

The ability of animals to sense and differentiate among thousands of odorants relies on a large set of olfactory receptors (OR) and a multitude of accessory proteins within the olfactory epithelium (OE). ORs and related signaling mechanisms have been the subject of intensive studies over the past years, but our knowledge regarding olfactory processing remains limited. The recent development of next generation sequencing (NGS) techniques encouraged us to assess the transcriptome of the murine OE. We analyzed RNA from OEs of female and male adult mice and from fluorescence-activated cell sorting (FACS)-sorted olfactory receptor neurons (ORNs) obtained from transgenic OMP-GFP mice. The Illumina RNA-Seq protocol was utilized to generate up to 86 million reads per transcriptome. In OE samples, nearly all OR and trace amine-associated receptor (TAAR) genes involved in the perception of volatile amines were detectably expressed. Other genes known to participate in olfactory signaling pathways were among the 200 genes with the highest expression levels in the OE. To identify OE-specific genes, we compared olfactory neuron expression profiles with RNA-Seq transcriptome data from different murine tissues. By analyzing different transcript classes, we detected the expression of non-olfactory GPCRs in ORNs and established an expression ranking for GPCRs detected in the OE. We also identified other previously undescribed membrane proteins as potential new players in olfaction. The quantitative and comprehensive transcriptome data provide a virtually complete catalogue of genes expressed in the OE and present a useful tool to uncover candidate genes involved in, for example, olfactory signaling, OR trafficking and recycling, and proliferation.


Subject(s)
Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Transcriptome/physiology , Animals , Female , Flow Cytometry , High-Throughput Nucleotide Sequencing , Male , Mice , Mice, Transgenic , Olfactory Pathways/metabolism , Signal Transduction/physiology
12.
Biochim Biophys Acta ; 1854(6): 632-40, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25219547

ABSTRACT

The prostate-specific G-protein-coupled receptor 1 (PSGR1) is an olfactory receptor specifically expressed in the prostate gland. PSGR1 expression is elevated both in benign prostatic hyperplasia tissue and in prostate cancer. Stimulation of PSGR1 by the odorant ß-ionone leads to an increase in the intracellular Ca(2+) concentration, activation of mitogen-activated protein (MAP) kinases and a decrease in prostate cancer cell proliferation. To further extend our knowledge about PSGR1 signaling in prostate cancer cells, we performed a quantitative phosphoproteomics study using stable isotope labeling by amino acids in cell culture and mass spectrometry. We report 51 differentially regulated phosphorylation sites in 24 proteins with functions in cytoskeletal remodeling, signaling and ion transport. Activation of PSGR1 evoked an increase in intracellular pH mediated by the sodium/hydrogen exchanger NHE1. Furthermore, we report the protein tyrosine kinase Pyk2 as a central effector of PSGR1 signaling cascades in LNCaP cells. Our data show that phosphorylation of p38 MAP kinase is triggered by Pyk2. In addition, we confirmed dephosphorylation of the tumor suppressor protein N-myc downstream regulated gene 1 (NDRG1) at Ser330 downstream of Pyk2. Since NDRG1 impacts oncogenic signaling pathways interfering with tumor progression, we suggest that the Pyk2-NDRG1 axis is possibly involved in conveying the anti-proliferative effect of ß-ionone in prostate cancer cells. This article is part of a Special Issue entitled: Medical Proteomics.


Subject(s)
Focal Adhesion Kinase 2/metabolism , MAP Kinase Signaling System , Neoplasm Proteins/metabolism , Phosphoproteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Odorant/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Focal Adhesion Kinase 2/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Neoplasm Proteins/genetics , Norisoprenoids/pharmacology , Phosphoproteins/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Prostatic Neoplasms/genetics , Receptors, Odorant/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Chem Senses ; 40(2): 73-87, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25500808

ABSTRACT

Vertebrates can sense and identify a vast array of chemical cues. The molecular machinery involved in chemodetection and transduction is expressed within the cilia of olfactory sensory neurons. Currently, there is only limited information available on the distribution and density of individual signaling components within the ciliary compartment. Using super-resolution microscopy, we show here that cyclic-nucleotide-gated channels and calcium-activated chloride channels of the anoctamin family are localized to discrete microdomains in the ciliary membrane. In addition to ANO2, a second anoctamin, ANO6, also localizes to ciliary microdomains. This observation, together with the fact that ANO6 and ANO2 co-localize, indicates a role for ANO6 in olfactory signaling. We show that both ANO2 and ANO6 can form heteromultimers and that this heteromerization alters the recombinant channels' physiological properties. Thus, we provide evidence for interaction of ANO2 and ANO6 in olfactory cilia, with possible physiological relevance for olfactory signaling.


Subject(s)
Chloride Channels/metabolism , Cilia/metabolism , Olfactory Mucosa/cytology , Phospholipid Transfer Proteins/metabolism , Sensory Receptor Cells/metabolism , Animals , Anoctamins , Chloride Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Mice, Inbred C57BL , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Phospholipid Transfer Proteins/genetics , Protein Multimerization , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
14.
J Cell Sci ; 127(Pt 11): 2518-27, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24652834

ABSTRACT

The olfactory signal transduction cascade transforms odor information into electrical signals by a cAMP-based amplification mechanism. The mechanisms underlying the very precise temporal and spatial organization of the relevant signaling components remains poorly understood. Here, we identify, using co-immunoprecipitation experiments, a macromolecular assembly of signal transduction components in mouse olfactory neurons, organized through MUPP1. Disruption of the PDZ signaling complex, through use of an inhibitory peptide, strongly impaired odor responses and changed the activation kinetics of olfactory sensory neurons. In addition, our experiments demonstrate that termination of the response is dependent on PDZ-based scaffolding. These findings provide new insights into the functional organization, and regulation, of olfactory signal transduction.


Subject(s)
Carrier Proteins/metabolism , Multiprotein Complexes/metabolism , Olfactory Mucosa/physiology , Animals , Carrier Proteins/genetics , Cyclic AMP/metabolism , HEK293 Cells , Humans , Membrane Proteins , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Olfactory Receptor Neurons/metabolism , PDZ Domains/genetics , Peptide Fragments/metabolism , Protein Binding , Receptors, Odorant/metabolism , Signal Transduction
15.
PLoS One ; 8(10): e77509, 2013.
Article in English | MEDLINE | ID: mdl-24204848

ABSTRACT

Various genetic or toxin-induced mouse models are frequently used for investigation of early PD pathology. Although olfactory impairment is known to precede motor symptoms by years, it is not known whether it is caused by impairments in the brain, the olfactory epithelium, or both. In this study, we investigated the olfactory function in three genetic Parkinson's disease (PD) mouse models and mice treated with MPTP intraperitoneally and intranasally. To investigate olfactory function, we performed electro-olfactogram recordings (EOGs) and an olfactory behavior test (cookie-finding test). We show that neither a parkin knockout mouse strain, nor intraperitoneal MPTP treated animals display any olfactory impairment in EOG recordings and the applied behavior test. We also found no difference in the responses of the olfactory epithelium to odorants in a mouse strain over-expressing doubly mutated α-synuclein, while this mouse strain was not suitable to test olfaction in a cookie-finding test as it displays a mobility impairment. A transgenic mouse expressing mutated α-synuclein in dopaminergic neurons performed equal to control animals in the cookie-finding test. Further we show that intranasal MPTP application can cause functional damage of the olfactory epithelium.


Subject(s)
Dopaminergic Neurons/metabolism , Olfactory Mucosa/metabolism , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/physiopathology , Smell/physiology , Ubiquitin-Protein Ligases/genetics , alpha-Synuclein/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Administration, Intranasal , Animals , Brain/metabolism , Brain/physiopathology , Discrimination Learning , Disease Models, Animal , Dopaminergic Neurons/pathology , Female , Gene Expression , Humans , Injections, Intraperitoneal , Male , Mice , Mutation , Odorants , Olfactory Mucosa/physiopathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Ubiquitin-Protein Ligases/deficiency , alpha-Synuclein/metabolism
16.
Chem Senses ; 38(3): 231-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23329732

ABSTRACT

Heteromeric insect odorant receptors (ORs) form ligand-activated nonselective cation channels in recombinant expression systems. We performed a pharmacological characterization of Drosophila melanogaster and Bombyx mori ORs expressed in the Xenopus laevis oocyte expression system and characterized them using the 2-electrode voltage clamp. We identified amiloride derivatives as high-affinity blockers, which inhibit the ion current through the channel in a low micromolar range. For the heteromeric Drosophila Or47a + DmelOrco receptor, the potency sequence (IC(50)) is HMA [5-(N,N-hexamethylene)amiloride] (3.9 µM), MIA [5-(N-methyl-N-isobutyl)amiloride] (11.0 µM), and DMA [5-(N,N-dimethyl)amiloride] (113.3 µM). Amiloride itself is nearly ineffective. Other tested insect ORs (Drosophila Or49b + DmelOrco, B. mori BmorOr1 + BmorOrco) were blocked in a similar fashion suggesting that the amiloride derivatives were potential general blockers of all receptor combinations. Our results suggest that pyrazine derivatives of amiloride are useful probes to study the mechanism of chemosensory transduction in insects in more detail.


Subject(s)
Amiloride/analogs & derivatives , Drosophila melanogaster/drug effects , Amiloride/pharmacology , Animals , Bombyx/drug effects , Bombyx/metabolism , Drosophila melanogaster/metabolism , Electrodes , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Pentanols/pharmacology , Receptors, Odorant/antagonists & inhibitors , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Xenopus laevis/growth & development , Xenopus laevis/metabolism
17.
Nat Neurosci ; 15(5): 754-62, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446879

ABSTRACT

In olfactory sensory neurons (OSNs), cytosolic Ca(2+) controls the gain and sensitivity of olfactory signaling. Important components of the molecular machinery that orchestrates OSN Ca(2+) dynamics have been described, but key details are still missing. Here, we demonstrate a critical physiological role of mitochondrial Ca(2+) mobilization in mouse OSNs. Combining a new mitochondrial Ca(2+) imaging approach with patch-clamp recordings, organelle mobility assays and ultrastructural analyses, our study identifies mitochondria as key determinants of olfactory signaling. We show that mitochondrial Ca(2+) mobilization during sensory stimulation shapes the cytosolic Ca(2+) response profile in OSNs, ensures a broad dynamic response range and maintains sensitivity of the spike generation machinery. When mitochondrial function is impaired, olfactory neurons function as simple stimulus detectors rather than as intensity encoders. Moreover, we describe activity-dependent recruitment of mitochondria to olfactory knobs, a mechanism that provides a context-dependent tool for OSNs to maintain cellular homeostasis and signaling integrity.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Olfactory Receptor Neurons/ultrastructure , Signal Transduction/physiology , Action Potentials/drug effects , Animals , Animals, Newborn , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Enzyme Inhibitors/pharmacology , Female , Fluorescence Recovery After Photobleaching/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Olfactory Bulb/cytology , Olfactory Receptor Neurons/metabolism , Organic Chemicals/pharmacokinetics , Patch-Clamp Techniques , Proton Ionophores/pharmacology , Receptors, Odorant/metabolism , Ruthenium Compounds/pharmacology , Signal Transduction/drug effects , Time Factors , Transfection
18.
Traffic ; 13(1): 120-30, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22008230

ABSTRACT

The role of actin, class I myosins and dynamin in endocytic uptake processes is well characterized, but their role during endo-phagosomal membrane trafficking and maturation is less clear. In Dictyostelium, knockout of myosin IB (myoB) leads to a defect in membrane protein recycling from endosomes back to the plasma membrane. Here, we show that actin plays a central role in the morphology and function of the endocytic pathway. Indeed, latrunculin B (LatB) induces endosome tubulation, a phenotype also observed in dynamin A (dymA)-null cells. Knockout of dymA impairs phagosome acidification, whereas knockout of myoB delays reneutralization, a phenotype mimicked by a low dose of LatB. As a read out for actin-dependent processes during maturation, we monitored the capacity of purified phagosomes to bind F-actin in vitro, and correlated this with the presence of actin-binding and membrane-trafficking proteins. Phagosomes isolated from myoB-null cells showed an increased binding to F-actin, especially late phagosomes. In contrast, early phagosomes from dymA-null cells showed reduced binding to F-actin while late phagosomes were unaffected. We provide evidence that Abp1 is the main F-actin-binding protein in this assay and is central for the interplay between DymA and MyoB during phagosome maturation.


Subject(s)
Actins/metabolism , Dynamins/metabolism , Endosomes/metabolism , Microfilament Proteins/metabolism , Myosin Type I/metabolism , Phagosomes/metabolism , Protozoan Proteins/metabolism , Blotting, Western , Dictyostelium/metabolism , Dictyostelium/ultrastructure , Dynamins/genetics , Endosomes/ultrastructure , Gene Knockout Techniques , Models, Biological , Myosin Type I/genetics , Phagocytosis , Phagosomes/ultrastructure , Protein Transport , Protozoan Proteins/genetics
20.
BMC Neurosci ; 12: 86, 2011 Aug 22.
Article in English | MEDLINE | ID: mdl-21859486

ABSTRACT

BACKGROUND: Extracellular nucleotides have long been known to play neuromodulatory roles and to be involved in intercellular signalling. In the olfactory system, ATP is released by olfactory neurons, and exogenous ATP can evoke an increase in intracellular calcium concentration in sustentacular cells, the nonneuronal supporting cells of the olfactory epithelium. Here we investigate the hypothesis that olfactory neurons communicate with sustentacular cells via extracellular ATP and purinergic receptor activation. RESULTS: Here we show that exposure of mice to a mixture of odorants induced a significant increase in the levels of the transcription factor CREB phosphorylated at Ser-133 in the nuclei of both olfactory sensory neurons and sustentacular cells. This activation was dependent on adenylyl cyclase III-mediated olfactory signaling and on activation of P2Y purinergic receptors on sustentacular cells. Purinergic receptor antagonists inhibited odorant-dependent CREB phosphorylation specifically in the nuclei of the sustentacular cells. CONCLUSION: Our results point to a possible role for extracellular nucleotides in mediating intercellular communication between the neurons and sustentacular cells of the olfactory epithelium in response to odorant exposure. Maintenance of extracellular ionic gradients and metabolism of noxious chemicals by sustentacular cells may therefore be regulated in an odorant-dependent manner by olfactory sensory neurons.


Subject(s)
CREB-Binding Protein/metabolism , Odorants , Olfactory Mucosa/physiology , Olfactory Receptor Neurons/physiology , Purinergic Antagonists/pharmacology , Receptors, Odorant/metabolism , Smell/physiology , Animals , Cells, Cultured , Mice , Olfactory Mucosa/cytology , Olfactory Mucosa/drug effects , Olfactory Receptor Neurons/drug effects , Phosphorylation , Smell/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...