1
|
Nakamuta S, Sakuma A, Nikaido M, Kato H, Miyazaki M, Yamamoto Y, Nakamuta N. Expression of type 1 vomeronasal receptors in the olfactory organ of the African lungfish, Protopterus dolloi. Acta Histochem 2023; 125:152078. [PMID: 37540956 DOI: 10.1016/j.acthis.2023.152078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/06/2023]
Abstract
The vomeronasal organ is an olfactory organ found in amphibians and higher vertebrates. Type 1 vomeronasal receptors, one of the major olfactory receptors in vertebrates, are expressed in the vomeronasal organ in mammals. In amphibians and fish, they are expressed in the olfactory epithelium. The lungfish, which is the species of fish most closely related to amphibians, has a primitive vomeronasal organ: the recess epithelium. Expression of type 1 vomeronasal receptors has been reported in both the olfactory epithelium and the recess epithelium in three species of African lungfish and one species of South American lungfish. However, a previous study suggested that in the African lungfish Protopterus dolloi these receptors are expressed only in the olfactory epithelium. In this study, we identified 21 type 1 vomeronasal receptor genes in P. dolloi and examined the expression sites in the olfactory organ. In P. dolloi, most cells expressing the type 1 vomeronasal receptor were distributed in the olfactory epithelium, but a few were also found in the recess epithelium. This implies that the functions of the olfactory epithelium and the primitive vomeronasal organ are incompletely separated, and that all extant African and South American lungfish share this trait.
Collapse
Affiliation(s)
- Shoko Nakamuta
- Laboratory of Veterinary Anatomy, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Atsuhiro Sakuma
- School of Life Science and Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8550, Japan
| | - Masato Nikaido
- School of Life Science and Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8550, Japan
| | - Hideaki Kato
- Faculty of Education, Shizuoka University, 836 Ohya, Shizuoka, 422-8529, Japan
| | - Masao Miyazaki
- Department of Biological Chemistry and Food Sciences, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Yoshio Yamamoto
- Laboratory of Veterinary Anatomy, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Nobuaki Nakamuta
- Laboratory of Veterinary Anatomy, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan.
| |
Collapse
|
2
|
Zhu KW, Burton SD, Nagai MH, Silverman JD, de March CA, Wachowiak M, Matsunami H. Decoding the olfactory map through targeted transcriptomics links murine olfactory receptors to glomeruli. Nat Commun 2022; 13:5137. [PMID: 36050313 PMCID: PMC9437035 DOI: 10.1038/s41467-022-32267-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/21/2022] [Indexed: 12/12/2022] Open
Abstract
Sensory processing in olfactory systems is organized across olfactory bulb glomeruli, wherein axons of peripheral sensory neurons expressing the same olfactory receptor co-terminate to transmit receptor-specific activity to central neurons. Understanding how receptors map to glomeruli is therefore critical to understanding olfaction. High-throughput spatial transcriptomics is a rapidly advancing field, but low-abundance olfactory receptor expression within glomeruli has previously precluded high-throughput mapping of receptors to glomeruli in the mouse. Here we combined sequential sectioning along the anteroposterior, dorsoventral, and mediolateral axes with target capture enrichment sequencing to overcome low-abundance target expression. This strategy allowed us to spatially map 86% of olfactory receptors across the olfactory bulb and uncover a relationship between OR sequence and glomerular position.
Collapse
Affiliation(s)
- Kevin W Zhu
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Shawn D Burton
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, 18015, USA
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Maira H Nagai
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Justin D Silverman
- College of Information Science and Technology, Pennsylvania State University, University Park, PA, 16802, USA
- Department of Statistics, Pennsylvania State University, University Park, PA, 16802, USA
- Department of Medicine, Pennsylvania State University, Hershey, PA, 17033, USA
- Institute for Computational and Data Science, Pennsylvania State University, University Park, PA, 16802, USA
| | - Claire A de March
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Matt Wachowiak
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA.
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Institute for Brain Sciences, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
3
|
Wang IH, Murray E, Andrews G, Jiang HC, Park SJ, Donnard E, Durán-Laforet V, Bear DM, Faust TE, Garber M, Baer CE, Schafer DP, Weng Z, Chen F, Macosko EZ, Greer PL. Spatial transcriptomic reconstruction of the mouse olfactory glomerular map suggests principles of odor processing. Nat Neurosci 2022; 25:484-492. [PMID: 35314823 PMCID: PMC9281876 DOI: 10.1038/s41593-022-01030-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 02/07/2022] [Indexed: 12/14/2022]
Abstract
The olfactory system's ability to detect and discriminate between the vast array of chemicals present in the environment is critical for an animal's survival. In mammals, the first step of this odor processing is executed by olfactory sensory neurons, which project their axons to a stereotyped location in the olfactory bulb (OB) to form glomeruli. The stereotyped positioning of glomeruli in the OB suggests an importance for this organization in odor perception. However, because the location of only a limited subset of glomeruli has been determined, it has been challenging to determine the relationship between glomerular location and odor discrimination. Using a combination of single-cell RNA sequencing, spatial transcriptomics and machine learning, we have generated a map of most glomerular positions in the mouse OB. These observations significantly extend earlier studies and suggest an overall organizational principle in the OB that may be used by the brain to assist in odor decoding.
Collapse
Affiliation(s)
- I-Hao Wang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Evan Murray
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Greg Andrews
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Hao-Ching Jiang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sung Jin Park
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Elisa Donnard
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Violeta Durán-Laforet
- Department of Neurobiology and Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Daniel M Bear
- Department of Psychology, Stanford University, Palo Alto, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Palo Alto, CA, USA
| | - Travis E Faust
- Department of Neurobiology and Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Manuel Garber
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christina E Baer
- Sanderson Center for Optical Imaging and Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dorothy P Schafer
- Department of Neurobiology and Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Evan Z Macosko
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Paul L Greer
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Bast WG, Albeanu DF. Mapping odorant receptors to their glomeruli. Nat Neurosci 2022; 25:405-407. [DOI: 10.1038/s41593-022-01045-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
5
|
Krull F, Hirschfeld M, Wemheuer WE, Brenig B. Frameshift Variant in Novel Adenosine-A1-Receptor Homolog Associated With Bovine Spastic Syndrome/Late-Onset Bovine Spastic Paresis in Holstein Sires. Front Genet 2020; 11:591794. [PMID: 33329738 PMCID: PMC7734149 DOI: 10.3389/fgene.2020.591794] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/26/2020] [Indexed: 11/13/2022] Open
Abstract
Since their first description almost 100 years ago, bovine spastic paresis (BSP) and bovine spastic syndrome (BSS) are assumed to be inherited neuronal-progressive diseases in cattle. Affected animals are characterized by (frequent) spasms primarily located in the hind limbs, accompanied by severe pain symptoms and reduced vigor, thus initiating premature slaughter or euthanasia. Due to the late onset of BSP and BSS and the massively decreased lifespan of modern cattle, the importance of these diseases is underestimated. In the present study, BSP/BSS-affected German Holstein breeding sires from artificial insemination centers were collected and pedigree analysis, genome-wide association studies, whole genome resequencing, protein-protein interaction network analysis, and protein-homology modeling were performed to elucidate the genetic background. The analysis of 46 affected and 213 control cattle revealed four significantly associated positions on chromosome 15 (BTA15), i.e., AC_000172.1:g.83465449A>G (-log10P = 19.17), AC_000172.1:g.81871849C>T (-log10P = 8.31), AC_000172.1:g.81872621A>T (-log10P = 6.81), and AC_000172.1:g.81872661G>C (-log10P = 6.42). Two additional loci were significantly associated located on BTA8 and BTA19, i.e., AC_000165.1:g.71177788T>C and AC_000176.1:g.30140977T>G, respectively. Whole genome resequencing of five affected individuals and six unaffected relatives (two fathers, two mothers, a half sibling, and a full sibling) belonging to three different not directly related families was performed. After filtering, a homozygous loss of function variant was identified in the affected cattle, causing a frameshift in the so far unknown gene locus LOC100848076 encoding an adenosine-A1-receptor homolog. An allele frequency of the variant of 0.74 was determined in 3,093 samples of the 1000 Bull Genomes Project.
Collapse
Affiliation(s)
- Frederik Krull
- Department of Animal Sciences, Faculty of Agricultural Sciences, Institute of Veterinary Medicine, University of Göttingen, Göttingen, Germany
| | - Marc Hirschfeld
- Department of Animal Sciences, Faculty of Agricultural Sciences, Institute of Veterinary Medicine, University of Göttingen, Göttingen, Germany
| | - Wilhelm Ewald Wemheuer
- Department of Animal Sciences, Faculty of Agricultural Sciences, Institute of Veterinary Medicine, University of Göttingen, Göttingen, Germany
| | - Bertram Brenig
- Department of Animal Sciences, Faculty of Agricultural Sciences, Institute of Veterinary Medicine, University of Göttingen, Göttingen, Germany
| |
Collapse
|
6
|
Baker KL, Vasan G, Gumaste A, Pieribone VA, Verhagen JV. Spatiotemporal dynamics of odor responses in the lateral and dorsal olfactory bulb. PLoS Biol 2019; 17:e3000409. [PMID: 31532763 PMCID: PMC6768483 DOI: 10.1371/journal.pbio.3000409] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 09/30/2019] [Accepted: 08/22/2019] [Indexed: 01/04/2023] Open
Abstract
The mammalian olfactory bulb (OB) plays an essential role in odor processing during the perception of smell. Optical imaging of the OB has proven to be a key tool in elucidating the spatial odor mapping and temporal dynamics that underlie higher-order odor processing. Much is known about the activation of olfactory sensory neuron (OSN) glomerular responses in the dorsal olfactory bulb (dOB) during odor presentation. However, the dorsal bulb provides access to only approximately 25% of all glomeruli, and little is known about how the lateral bulb functions during this critical process. Here, we report, for the first time, simultaneous measurements of OSN glomerular activity from both the dOB and the lateral olfactory bulb (lOB), thus describing odor-specific spatial mapping and the temporal dynamics of olfactory input to both the dorsal and lateral bulb. Odor responses in the lateral bulb tended to be most prominent in the dorso-lateral (D-L) region. Lateral glomeruli became active in a dorso-ventral (D-V) sequence upon odor inhalation, unlike the anterio-posterior (A-P) activity wave typical of the dorsal glomeruli. Across the entire D-L bulb, the spatial organization of these dynamics can be explained neither by the purely mechanosensitive dynamics (to breathing clean air) nor by the response amplitudes across glomeruli. Instead, these dynamics can be explained by a combination of zonal receptor distributions, associated OB projections, and air flow paths across the epithelium upon inhalation. Remarkably, we also found that a subset of OSN glomeruli in the lOB was highly sensitive to extranasal air pressure changes, a response type that has not been reported in dorsal glomeruli. The mammalian olfactory bulb plays an essential role in odor processing during the perception of smell, but most studies have focused on the dorsal olfactory bulb, which contains only a quarter of all glomeruli. In this study, imaging of the lateral olfactory bulb reveals new properties in smell processing.
Collapse
Affiliation(s)
- Keeley L. Baker
- The John B. Pierce Laboratory, New Haven, Connecticut, United States of America
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Ganesh Vasan
- The John B. Pierce Laboratory, New Haven, Connecticut, United States of America
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Ankita Gumaste
- The John B. Pierce Laboratory, New Haven, Connecticut, United States of America
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Vincent A. Pieribone
- The John B. Pierce Laboratory, New Haven, Connecticut, United States of America
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Justus V. Verhagen
- The John B. Pierce Laboratory, New Haven, Connecticut, United States of America
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
7
|
Inokuchi K, Imamura F, Takeuchi H, Kim R, Okuno H, Nishizumi H, Bito H, Kikusui T, Sakano H. Nrp2 is sufficient to instruct circuit formation of mitral-cells to mediate odour-induced attractive social responses. Nat Commun 2017; 8:15977. [PMID: 28731029 PMCID: PMC5525001 DOI: 10.1038/ncomms15977] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 05/16/2017] [Indexed: 01/22/2023] Open
Abstract
Odour information induces various innate responses that are critical to the survival of the individual and for the species. An axon guidance molecule, Neuropilin 2 (Nrp2), is known to mediate targeting of olfactory sensory neurons (primary neurons), to the posteroventral main olfactory bulb (PV MOB) in mice. Here we report that Nrp2-positive (Nrp2+) mitral cells (MCs, second-order neurons) play crucial roles in transmitting attractive social signals from the PV MOB to the anterior part of medial amygdala (MeA). Semaphorin 3F, a repulsive ligand to Nrp2, regulates both migration of Nrp2+ MCs to the PV MOB and their axonal projection to the anterior MeA. In the MC-specific Nrp2 knockout mice, circuit formation of Nrp2+ MCs and odour-induced attractive social responses are impaired. In utero, electroporation demonstrates that activation of the Nrp2 gene in MCs is sufficient to instruct their circuit formation from the PV MOB to the anterior MeA.
Collapse
Affiliation(s)
- Kasumi Inokuchi
- Department of Brain Function, School of Medical Science, University of Fukui, Fukui 910-1193, Japan.,Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
| | - Fumiaki Imamura
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Haruki Takeuchi
- Department of Brain Function, School of Medical Science, University of Fukui, Fukui 910-1193, Japan
| | - Ryang Kim
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroyuki Okuno
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Hirofumi Nishizumi
- Department of Brain Function, School of Medical Science, University of Fukui, Fukui 910-1193, Japan.,Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Takefumi Kikusui
- Department of Animal Science and Biotechnology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa 252-5201, Japan
| | - Hitoshi Sakano
- Department of Brain Function, School of Medical Science, University of Fukui, Fukui 910-1193, Japan
| |
Collapse
|
8
|
Low VF, Mombaerts P. Odorant receptor proteins in the mouse main olfactory epithelium and olfactory bulb. Neuroscience 2017; 344:167-177. [PMID: 28057532 DOI: 10.1016/j.neuroscience.2016.12.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 01/10/2023]
Abstract
In the mouse, odorant receptor proteins (ORs) are G-protein-coupled receptors expressed in mature olfactory sensory neurons (OSNs) of the main olfactory epithelium (MOE). ORs mediate odorant reception at the level of the OSN cilia. Most of the ∼1100 OR genes in the mouse genome are expressed, at the RNA level, in mature OSNs. The literature on antibodies against ORs is limited, and most reports are with antibodies that are not commercially available. Here we have screened 40 commercial antibodies against human and mouse ORs by immunofluorescence staining of coronal cryosections of the MOE of 21-day-old C57BL/6J mice. Various methods of antigen retrieval were tested. Of the 19 antibodies raised against human ORs, three yielded a consistent immunoreactive signal in the mouse MOE; of these three, two appeared to cross react against one or more, unknown, mouse ORs. Of the 21 antibodies raised against mouse ORs, six yielded a consistent immunoreactive signal in the mouse MOE; of these six, two also stained specific glomeruli in the olfactory bulb. Antibody specificity could be validated with gene-targeted mouse strains in the case of three ORs. The number of OSNs immunoreactive for the MOR28/Olfr1507 antibody is greater in C57BL/6J than in 129S6/SvEvTac wild-type mice. Taken together, our results are encouraging: 20-30% of these commercially available antibodies are informative in immunohistochemical analyses of the mouse MOE. The commercial availability of these antibodies should facilitate the study of OR proteins in the MOE and the olfactory bulb, and the replicability of results in the literature.
Collapse
Affiliation(s)
- Victoria F Low
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, D-60438 Frankfurt, Germany.
| | - Peter Mombaerts
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, D-60438 Frankfurt, Germany.
| |
Collapse
|
9
|
CD36 is expressed in a defined subpopulation of neurons in the olfactory epithelium. Sci Rep 2016; 6:25507. [PMID: 27145700 PMCID: PMC4857125 DOI: 10.1038/srep25507] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 04/19/2016] [Indexed: 11/08/2022] Open
Abstract
The sensory neurons in the olfactory epithelium (OSNs) are equipped with a large repertoire of olfactory receptors and the associated signal transduction machinery. In addition to the canonical OSNs, which express odorant receptors (ORs), the epithelium contains specialized subpopulations of sensory neurons that can detect specific information from environmental cues and relay it to relevant neuronal circuitries. Here we describe a subpopulation of mature OSNs in the main olfactory epithelium (MOE) which expresses CD36, a multifunctional receptor involved in a series of biological processes, including sensory perception of lipid ligands. The Cd36 expressing neurons coexpress markers of mature OSNs and are dispersed throughout the MOE. Unlike several ORs analyzed in our study, we found frequent coexpression of the OR Olfr287 in these neurons, suggesting that only a specific set of ORs may be coexpressed with CD36 in OSNs. We also show that CD36 is expressed in the cilia of OSNs, indicating a possible role in odorant detection. CD36-deficient mice display no signs of gross changes in the organization of the olfactory epithelium, but show impaired preference for a lipid mixture odor. Our results show that CD36-expressing neurons represent a distinct population of OSNs, which may have specific functions in olfaction.
Collapse
|
10
|
Multiplex assessment of the positions of odorant receptor-specific glomeruli in the mouse olfactory bulb by serial two-photon tomography. Proc Natl Acad Sci U S A 2015; 112:E5873-82. [PMID: 26450880 DOI: 10.1073/pnas.1512135112] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the mouse, axons of olfactory sensory neurons (OSNs) that express the same odorant receptor (OR) gene coalesce into one or a few glomeruli in the olfactory bulb. The positions of OR-specific glomeruli are traditionally described as stereotyped. Here, we have assessed quantitatively the positions of OR-specific glomeruli using serial two-photon tomography, an automated method for whole-organ fluorescence imaging that integrates two-photon microscopy with serial microtome sectioning. Our strategy is multiplexed. By repeated crossing, we generated two strains of mice with gene-targeted mutations at four or five OR loci for a total of six ORs: MOR23 (Olfr16), mOR37A (Olfr155), M72 (Olfr160), P2 (Olfr17), MOR256-17 (Olfr15), and MOR28 (Olfr1507). Glomerular imaging relied on intrinsic fluorescence of GFP or DsRed, or on whole-mount immunofluorescence with antibodies against GFP, DsRed, or β-gal using the method of immunolabeling-enabled three-dimensional imaging of solvent-cleared organs (iDISCO). The high-resolution 3D-reconstructed datasets were segmented to identify the labeled glomeruli and to assess glomerular positional variability between the bulbs of one mouse (intraindividual) and among the bulbs of different mice (interindividual). In 26 mice aged 21 or 50 d or 10 wk, we made measurements of the positions of 352 glomeruli. We find that positional variability of glomeruli correlates with the OR: For instance, the medial MOR28 glomerular domain occupies a surface area that is an order of magnitude larger than the surface area of the medial MOR23 glomerular domain. Our results quantify the level of precision that is delivered by the mechanisms of OSN axon wiring, differentially for the various OSN populations expressing distinct OR genes.
Collapse
|
11
|
Supersensitive detection and discrimination of enantiomers by dorsal olfactory receptors: evidence for hierarchical odour coding. Sci Rep 2015; 5:14073. [PMID: 26361056 PMCID: PMC4566093 DOI: 10.1038/srep14073] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/18/2015] [Indexed: 11/08/2022] Open
Abstract
Enantiomeric pairs of mirror-image molecular structures are difficult to resolve by instrumental analyses. The human olfactory system, however, discriminates (−)-wine lactone from its (+)-form rapidly within seconds. To gain insight into receptor coding of enantiomers, we compared behavioural detection and discrimination thresholds of wild-type mice with those of ΔD mice in which all dorsal olfactory receptors are genetically ablated. Surprisingly, wild-type mice displayed an exquisite “supersensitivity” to enantiomeric pairs of wine lactones and carvones. They were capable of supersensitive discrimination of enantiomers, consistent with their high detection sensitivity. In contrast, ΔD mice showed selective major loss of sensitivity to the (+)-enantiomers. The resulting 108-fold differential sensitivity of ΔD mice to (−)- vs. (+)-wine lactone matched that observed in humans. This suggests that humans lack highly sensitive orthologous dorsal receptors for the (+)-enantiomer, similarly to ΔD mice. Moreover, ΔD mice showed >1010-fold reductions in enantiomer discrimination sensitivity compared to wild-type mice. ΔD mice detected one or both of the (−)- and (+)-enantiomers over a wide concentration range, but were unable to discriminate them. This “enantiomer odour discrimination paradox” indicates that the most sensitive dorsal receptors play a critical role in hierarchical odour coding for enantiomer identification.
Collapse
|
12
|
Odorant receptors signaling instructs the development and plasticity of the glomerular map. Neural Plast 2015; 2015:975367. [PMID: 25688305 PMCID: PMC4320882 DOI: 10.1155/2015/975367] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/24/2014] [Accepted: 12/24/2014] [Indexed: 12/21/2022] Open
Abstract
The olfactory system provides a great opportunity to explore the mechanisms that underlie the formation and function of neural circuits because of the simplicity of its structure. Olfactory sensory neurons (OSNs) located in the peripheral olfactory epithelium (OE) take part in the initial formation and function of glomeruli in the olfactory bulb (OB) inside the central nervous system. Glomeruli are key in the process of transduction of olfactory information, as they constitute a map in the OB that sorts the different types of odorant inputs. This odorant categorization allows proper olfactory perception, and it is achieved through the anatomical organization and function of the different glomerular circuits. Once formed, glomeruli keep the capacity to undergo diverse plasticity processes, which is unique among the different neural circuits of the central nervous system. In this context, through the expression and function of the odorant receptors (ORs), OSNs perform two of the most important roles in the olfactory system: transducing odorant information to the nervous system and initiating the development of the glomerular map to organize olfactory information. This review addresses essential information that has emerged in recent years about the molecular basis of these processes.
Collapse
|
13
|
Yoshihara SI, Takahashi H, Nishimura N, Kinoshita M, Asahina R, Kitsuki M, Tatsumi K, Furukawa-Hibi Y, Hirai H, Nagai T, Yamada K, Tsuboi A. Npas4 Regulates Mdm2 and thus Dcx in Experience-Dependent Dendritic Spine Development of Newborn Olfactory Bulb Interneurons. Cell Rep 2014; 8:843-57. [DOI: 10.1016/j.celrep.2014.06.056] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 06/05/2014] [Accepted: 06/26/2014] [Indexed: 02/03/2023] Open
|
14
|
Tsai L, Barnea G. A critical period defined by axon-targeting mechanisms in the murine olfactory bulb. Science 2014; 344:197-200. [PMID: 24723611 DOI: 10.1126/science.1248806] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The olfactory system remains plastic throughout life because of continuous neurogenesis of sensory neurons in the nose and inhibitory interneurons in the olfactory bulb. Here, we reveal that transgenic expression of an odorant receptor has non-cell autonomous effects on axons expressing this receptor from the endogenous gene. Perinatal expression of transgenic odorant receptor causes rerouting of like axons to new glomeruli, whereas expression after the sensory map is established does not lead to rerouting. Further, chemical ablation of the map after rerouting does not restore the normal map, even when the transgenic receptor is no longer expressed. Our results reveal that glomeruli are designated as targets for sensory neurons expressing specific odorant receptors during a critical period in the formation of the olfactory sensory map.
Collapse
Affiliation(s)
- Lulu Tsai
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | | |
Collapse
|
15
|
Neural map formation in the mouse olfactory system. Cell Mol Life Sci 2014; 71:3049-57. [PMID: 24638094 PMCID: PMC4111858 DOI: 10.1007/s00018-014-1597-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 02/26/2014] [Accepted: 02/27/2014] [Indexed: 01/19/2023]
Abstract
In the mouse olfactory system, odorants are detected by ~1,000 different odorant receptors (ORs) produced by olfactory sensory neurons (OSNs). Each OSN expresses only one functional OR species, which is referred to as the “one neuron–one receptor” rule. Furthermore, OSN axons bearing the same OR converge to a specific projection site in the olfactory bulb (OB) forming a glomerular structure, i.e., the “one glomerulus–one receptor” rule. Based on these basic rules, binding signals of odorants detected by OSNs are converted to topographic information of activated glomeruli in the OB. During development, the glomerular map is formed by the combination of two genetically programmed processes: one is OR-independent projection along the dorsal–ventral axis, and the other is OR-dependent projection along the anterior-posterior axis. The map is further refined in an activity-dependent manner during the neonatal period. Here, we summarize recent progress of neural map formation in the mouse olfactory system.
Collapse
|
16
|
Woods HA. Mosaic physiology from developmental noise: within-organism physiological diversity as an alternative to phenotypic plasticity and phenotypic flexibility. J Exp Biol 2014; 217:35-45. [DOI: 10.1242/jeb.089698] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A key problem in organismal biology is to explain the origins of functional diversity. In the context of organismal biology, functional diversity describes the set of phenotypes, across scales of biological organization and through time, that a single genotype, or genome, or organism, can produce. Functional diversity encompasses many phenomena: differences in cell types within organisms; physiological and morphological differences among tissues and organs; differences in performance; morphological shifts in external phenotype; and changes in behavior. How can single genomes produce so many different phenotypes? Modern biology proposes two general mechanisms. The first is developmental programs, by which single cells and their single genomes diversify, via relatively deterministic processes, into the sets of cell types, tissues and organs that we see in most multicellular organisms. The second general mechanism is phenotypic modification stemming from interactions between organisms and their environments – modifications known either as phenotypic plasticity or as phenotypic flexibility, depending on the time scale of the response and the degree of reversibility. These two diversity-generating mechanisms are related because phenotypic modifications may sometimes arise as a consequence of environments influencing developmental programs. Here, I propose that functional diversity also arises via a third fundamental mechanism: stochastic developmental events giving rise to mosaics of physiological diversity within individual organisms. In biological systems, stochasticity stems from the inherently random actions of small numbers of molecules interacting with one another. Although stochastic effects occur in many biological contexts, available evidence suggests that they can be especially important in gene networks, specifically as a consequence of low transcript numbers in individual cells. I briefly review known mechanisms by which organisms control such stochasticity, and how they may use it to create adaptive functional diversity. I then fold this idea into modern thinking on phenotypic plasticity and flexibility, proposing that multicellular organisms exhibit ‘mosaic physiology’. Mosaic physiology refers to sets of diversified phenotypes, within individual organisms, that carry out related functions at the same time, but that are distributed in space. Mosaic physiology arises from stochasticity-driven differentiation of cells, early during cell diversification, which is then amplified by cell division and growth into macroscopic phenotypic modules (cells, tissues, organs) making up the physiological systems of later life stages. Mosaic physiology provides a set of standing, diversified phenotypes, within single organisms, that raise the likelihood of the organism coping well with novel environmental challenges. These diversified phenotypes can be distinct, akin to polyphenisms at the organismal level; or they can be continuously distributed, creating a kind of standing, simultaneously expressed reaction norm of physiological capacities.
Collapse
Affiliation(s)
- H. Arthur Woods
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
17
|
Richard M, Jamet S, Fouquet C, Dubacq C, Boggetto N, Pincet F, Gourier C, Trembleau A. Homotypic and heterotypic adhesion induced by odorant receptors and the β2-adrenergic receptor. PLoS One 2013; 8:e80100. [PMID: 24312457 PMCID: PMC3846556 DOI: 10.1371/journal.pone.0080100] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 10/08/2013] [Indexed: 11/25/2022] Open
Abstract
In the mouse olfactory system regulated expression of a large family of G Protein-Coupled Receptors (GPCRs), the Odorant Receptors (ORs), provides each sensory neuron with a single OR identity. In the wiring of the olfactory sensory neuron projections, a complex axon sorting process ensures the segregation of >1,000 subpopulations of axons of the same OR identity into homogeneously innervated glomeruli. ORs are critical determinants in axon sorting, and their presence on olfactory axons raises the intriguing possibility that they may participate in axonal wiring through direct or indirect trans-interactions mediating adhesion or repulsion between axons. In the present work, we used a biophysical assay to test the capacity of ORs to induce adhesion of cell doublets overexpressing these receptors. We also tested the β2 Adrenergic Receptor, a non-OR GPCR known to recapitulate the functions of ORs in olfactory axon sorting. We report here the first evidence for homo- and heterotypic adhesion between cells overexpressing the ORs MOR256-17 or M71, supporting the hypothesis that ORs may contribute to olfactory axon sorting by mediating differential adhesion between axons.
Collapse
Affiliation(s)
- Marion Richard
- CNRS UMR 7102, Université Pierre et Marie Curie Paris 06, Team Development and Plasticity of Neural Networks, Paris, France
| | - Sophie Jamet
- CNRS UMR 7102, Université Pierre et Marie Curie Paris 06, Team Development and Plasticity of Neural Networks, Paris, France
| | - Coralie Fouquet
- CNRS UMR 7102, Université Pierre et Marie Curie Paris 06, Team Development and Plasticity of Neural Networks, Paris, France
| | - Caroline Dubacq
- CNRS UMR 7102, Université Pierre et Marie Curie Paris 06, Team Development and Plasticity of Neural Networks, Paris, France
| | - Nicole Boggetto
- Imaging Facility (ImagoSeine), Jacques Monod Institute, CNRS-Université Paris Diderot, Paris, France
| | - Frédéric Pincet
- Laboratoire de Physique Statistique, Ecole Normale Supérieure, UPMC Univ Paris 06, Université Paris Diderot, CNRS, Paris, France
| | - Christine Gourier
- Laboratoire de Physique Statistique, Ecole Normale Supérieure, UPMC Univ Paris 06, Université Paris Diderot, CNRS, Paris, France
| | - Alain Trembleau
- CNRS UMR 7102, Université Pierre et Marie Curie Paris 06, Team Development and Plasticity of Neural Networks, Paris, France
- * E-mail:
| |
Collapse
|
18
|
Aoki M, Takeuchi H, Nakashima A, Nishizumi H, Sakano H. Possible roles of Robo1+ ensheathing cells in guiding dorsal-zone olfactory sensory neurons in mouse. Dev Neurobiol 2013; 73:828-40. [PMID: 23821580 DOI: 10.1002/dneu.22103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 06/23/2013] [Accepted: 06/24/2013] [Indexed: 11/06/2022]
Abstract
In the mouse olfactory system, the anatomical locations of olfactory sensory neurons (OSNs) correlate with their axonal projection sites along the dorsoventral axis of the olfactory bulb (OB). We have previously reported that Neuropilin-2 expressed by ventral-zone OSNs contributes to the segregation of dorsal and ventral OSN axons, and that Slit is acting as a negative land mark to restrict the projection of Robo2+, early-arriving OSN axons to the embryonic OB. Here, we report that another guidance receptor, Robo1, also plays an important role in guiding OSN axons. Knockout mice for Robo1 demonstrated defects in targeting of OSN axons to the OB. Although Robo1 is colocalized with dorsal-zone OSN axons, it is not produced by OSNs, but instead by olfactory ensheathing cells. These findings indicate a novel strategy of axon guidance in the mouse olfactory system during development.
Collapse
Affiliation(s)
- Mari Aoki
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Tokyo, 113-0032, Japan
| | | | | | | | | |
Collapse
|
19
|
Nakashima A, Takeuchi H, Imai T, Saito H, Kiyonari H, Abe T, Chen M, Weinstein LS, Yu CR, Storm DR, Nishizumi H, Sakano H. Agonist-independent GPCR activity regulates anterior-posterior targeting of olfactory sensory neurons. Cell 2013; 154:1314-25. [PMID: 24034253 PMCID: PMC7394037 DOI: 10.1016/j.cell.2013.08.033] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 07/11/2013] [Accepted: 08/16/2013] [Indexed: 10/26/2022]
Abstract
G-protein-coupled receptors (GPCRs) are known to possess two different conformations, active and inactive, and they spontaneously alternate between the two in the absence of ligands. Here, we analyzed the agonist-independent GPCR activity for its possible role in receptor-instructed axonal projection. We generated transgenic mice expressing activity mutants of the β2-adrenergic receptor, a well-characterized GPCR with the highest homology to odorant receptors (ORs). We found that mutants with altered agonist-independent activity changed the transcription levels of axon-targeting molecules--e.g., Neuropilin-1 and Plexin-A1--but not of glomerular segregation molecules--e.g., Kirrel2 and Kirrel3--thus causing shifts in glomerular locations along the anterior-posterior (A-P) axis. Knockout and in vitro experiments demonstrated that Gs, but not Golf, is responsible for mediating the agonist-independent GPCR activity. We conclude that the equilibrium of conformational transitions set by each OR is the major determinant of expression levels of A-P-targeting molecules.
Collapse
Affiliation(s)
- Ai Nakashima
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
- Department of Brain Function, School of Medical Science, University of Fukui, Fukui 910-1193, Japan
- These authors contributed equally to this work
| | - Haruki Takeuchi
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
- Department of Brain Function, School of Medical Science, University of Fukui, Fukui 910-1193, Japan
- These authors contributed equally to this work
| | - Takeshi Imai
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
- Laboratory for Sensory Circuit Formation, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
- PRESTO, Japan Science and Technology Agency, Saitama 332-0012, Japan
- These authors contributed equally to this work
| | - Harumi Saito
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Min Chen
- Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Lee S. Weinstein
- Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | - Daniel R. Storm
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA
| | - Hirofumi Nishizumi
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
| | - Hitoshi Sakano
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
- Department of Brain Function, School of Medical Science, University of Fukui, Fukui 910-1193, Japan
| |
Collapse
|
20
|
Khan M, Vaes E, Mombaerts P. Temporal patterns of odorant receptor gene expression in adult and aged mice. Mol Cell Neurosci 2013; 57:120-9. [PMID: 23962816 DOI: 10.1016/j.mcn.2013.08.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 08/05/2013] [Accepted: 08/09/2013] [Indexed: 01/27/2023] Open
Abstract
In the mouse, the sense of smell relies predominantly on the expression of ~1200 odorant receptor (OR) genes in the main olfactory epithelium (MOE). Each mature olfactory sensory neuron (OSN) in the MOE is thought to express just one of these OR genes; conversely, an OR gene is expressed in thousands to tens of thousands of OSNs per mouse. Here, we have characterized temporal patterns of OR gene expression in a cohort of inbred C57BL6/N mice from the Aged Rodent Colonies of the National Institute on Aging. We applied the NanoString multiplex platform to quantify RNA abundance for 531 OR genes in whole olfactory mucosa (WOM) tissue samples. The five study groups were females aged 2, 6, 12, 18, and 31 months (mo). We classified the 531 temporal patterns using a step-down quadratic regression method for time course analysis. The majority of OR genes (58.4%) are classified as flat: there is no significant difference from a horizontal line within this time window. There are 32.8% of OR genes with a downward profile, 7.2% with an upward profile, and 1.7% with a convex or concave profile. But the magnitude of these decreases and increases tends to be small: only 4.3% of OR genes are differentially expressed (DE) at 31 mo compared to 2 mo. Interestingly, the variances of NanoString counts for individual OR genes are homogeneous among the age groups. Our analyses of these 15,930 OR gene expression data of C57BL6/N mice that were raised and housed under well-controlled conditions indicate that OR gene expression at the MOE level is intrinsically stable.
Collapse
Affiliation(s)
- Mona Khan
- Max Planck Research Unit for Molecular Neurogenetics, 60438 Frankfurt, Germany
| | | | | |
Collapse
|
21
|
Zhao S, Tian H, Ma L, Yuan Y, Yu CR, Ma M. Activity-dependent modulation of odorant receptor gene expression in the mouse olfactory epithelium. PLoS One 2013; 8:e69862. [PMID: 23922828 PMCID: PMC3726745 DOI: 10.1371/journal.pone.0069862] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 06/12/2013] [Indexed: 01/15/2023] Open
Abstract
Activity plays critical roles in development and maintenance of the olfactory system, which undergoes considerable neurogenesis throughout life. In the mouse olfactory epithelium, each olfactory sensory neuron (OSN) stably expresses a single odorant receptor (OR) type out of a repertoire of ∼1200 and the OSNs with the same OR identity are distributed within one of the few broadly-defined zones. However, it remains elusive whether and how activity modulates such OR expression patterns. Here we addressed this question by investigating OR gene expression via in situ hybridization when sensory experience or neuronal excitability is manipulated. We first examined the expression patterns of fifteen OR genes in mice which underwent neonatal, unilateral naris closure. After four-week occlusion, the cell density in the closed (sensory-deprived) side was significantly lower (for four ORs), similar (for three ORs), or significantly higher (for eight ORs) as compared to that in the open (over-stimulated) side, suggesting that sensory inputs have differential effects on OSNs expressing different OR genes. We next examined the expression patterns of seven OR genes in transgenic mice in which mature OSNs had reduced neuronal excitability. Neuronal silencing led to a significant reduction in the cell density for most OR genes tested and thinner olfactory epithelium with an increased density of apoptotic cells. These results suggest that sensory experience plays important roles in shaping OR gene expression patterns and the neuronal activity is critical for survival of OSNs.
Collapse
Affiliation(s)
- Shaohua Zhao
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Geriatric Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huikai Tian
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Limei Ma
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Ying Yuan
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Minghong Ma
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
22
|
Fuss SH, Zhu Y, Mombaerts P. Odorant receptor gene choice and axonal wiring in mice with deletion mutations in the odorant receptor gene SR1. Mol Cell Neurosci 2013; 56:212-24. [PMID: 23688909 DOI: 10.1016/j.mcn.2013.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/16/2013] [Accepted: 05/02/2013] [Indexed: 01/17/2023] Open
Abstract
In the mouse, a mature olfactory sensory neuron (OSN) of the main olfactory epithelium (MOE) expresses one allele of one of the 1200 odorant receptor (OR) genes in the genome. The mechanisms that underlie the one receptor-one neuron rule remain poorly understood. A popular experimental paradigm for OR gene choice is to delete an OR coding region by gene targeting or in a transgene. Here we have applied this ∆OR paradigm to SR1, also known as MOR256-3 or Olfr124. This gene is expressed in OSNs of the MOE, and in ~50% of the OSNs of the septal organ. In heterozygous ∆SR1 mice, we observe an unprecedented biallelic expression rate of 30% at the SR1 locus. In homozygous ∆SR1 mice, we find a significant increase in the number of septal organ OSNs that undergo apoptosis. As a population, ∆SR1 OSNs project their axons to 81-85 glomeruli in each half of the OB, and coexpress at least 77 OR genes as evaluated by single-cell molecular analysis. There are no obvious or simple rules for the set of OR genes that are coexpressed with the ∆SR1 allele. The frequencies of coexpression are different for ∆SR1 OSNs in the septal organ compared to those in the MOE. We propose that there are as many as five scenarios for the fate of individual ∆SR1 OSNs.
Collapse
Affiliation(s)
- Stefan H Fuss
- The Rockefeller University, New York, NY 10065, USA; Bogazici University, Department of Molecular Biology and Genetics, 34342 Bebek, Istanbul, Turkey
| | | | | |
Collapse
|
23
|
Lodovichi C, Belluscio L. Odorant receptors in the formation of the olfactory bulb circuitry. Physiology (Bethesda) 2012; 27:200-12. [PMID: 22875451 DOI: 10.1152/physiol.00015.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In mammals, smell is mediated by odorant receptors expressed by sensory neurons in the nose. These specialized receptors are found both on olfactory sensory neurons' cilia and axon terminals. Although the primary function of ciliary odorant receptors is to detect odorants, their axonal role remains unclear but is thought to involve axon guidance. This review discusses findings that show axonal odorant receptors are indeed functional and capable of modulating neural connectivity.
Collapse
Affiliation(s)
- Claudia Lodovichi
- Venetian Institute of Molecular Medicine, and Institute of Neuroscience-CNR, Padua, Italy
| | | |
Collapse
|
24
|
Cho JH, Kam JWK, Cloutier JF. Slits and Robo-2 regulate the coalescence of subsets of olfactory sensory neuron axons within the ventral region of the olfactory bulb. Dev Biol 2012; 371:269-79. [PMID: 22981605 DOI: 10.1016/j.ydbio.2012.08.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Revised: 08/22/2012] [Accepted: 08/23/2012] [Indexed: 11/16/2022]
Abstract
Olfactory sensory neurons (OSNs) project their axons to second-order neurons in the olfactory bulb (OB) to form a precise glomerular map and these stereotypic connections are crucial for accurate odorant information processing by animals. To form these connections, olfactory sensory neuron (OSN) axons respond to axon guidance molecules that direct their growth and coalescence. We have previously implicated the axon guidance receptor Robo-2 in the accurate coalescence of OSN axons within the dorsal region of the OB (Cho et al., 2011). Herein, we have examined whether Robo-2 and its ligands, the Slits, contribute to the formation of an accurate glomerular map within more ventral regions of the OB. We have ablated expression of Robo-2 in OSNs and assessed the targeting accuracy of axons expressing either the P2 or MOR28 olfactory receptors, which innervate two different regions of the ventral OB. We show that P2-positive axons, which express Robo-2, coalesce into glomeruli more ventrally and form additional glomeruli in the OB of robo-2(lox/lox);OMP-Cre mice. We also demonstrate that Robo-2-mediated targeting of P2 axons along the dorsoventral axis of the OB is controlled by Slit-1 and Slit-3 expression. Interestingly, although MOR28-positive OSNs only express low levels of Robo-2, a reduced number of MOR28-positive glomeruli is observed in the OB of robo-2(lox/lox);OMP-Cre mice. Taken together, our results demonstrate that Slits and Robo-2 are required for the formation of an accurate glomerular map in the ventral region of the OB.
Collapse
Affiliation(s)
- Jin H Cho
- Montreal Neurological Institute, Centre for Neuronal Survival, 3801 University, Montréal, Québec, Canada H3A 2B4
| | | | | |
Collapse
|
25
|
Zhang J, Huang G, Dewan A, Feinstein P, Bozza T. Uncoupling stimulus specificity and glomerular position in the mouse olfactory system. Mol Cell Neurosci 2012; 51:79-88. [PMID: 22926192 DOI: 10.1016/j.mcn.2012.08.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 08/10/2012] [Indexed: 11/28/2022] Open
Abstract
Sensory information is often mapped systematically in the brain with neighboring neurons responding to similar stimulus features. The olfactory system represents chemical information as spatial and temporal activity patterns across glomeruli in the olfactory bulb. However, the degree to which chemical features are mapped systematically in the glomerular array has remained controversial. Here, we test the hypothesis that the dual roles of odorant receptors, in axon guidance and odor detection, can serve as a mechanism to map olfactory inputs with respect to their function. We compared the relationship between response specificity and glomerular position in genetically-defined olfactory sensory neurons expressing variant odorant receptors. We find that sensory neurons with the same odor response profile can be mapped to different regions of the bulb, and that neurons with different response profiles can be mapped to the same glomeruli. Our data demonstrate that the two functions of odorant receptors can be uncoupled, indicating that the mechanisms that map olfactory sensory inputs to glomeruli do so without regard to stimulus specificity.
Collapse
Affiliation(s)
- Jingji Zhang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | | | | | | | | |
Collapse
|
26
|
Young JM, Luche RM, Trask BJ. Rigorous and thorough bioinformatic analyses of olfactory receptor promoters confirm enrichment of O/E and homeodomain binding sites but reveal no new common motifs. BMC Genomics 2011; 12:561. [PMID: 22085861 PMCID: PMC3247239 DOI: 10.1186/1471-2164-12-561] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 11/15/2011] [Indexed: 12/02/2022] Open
Abstract
Background Mammalian olfactory receptors (ORs) are subject to a remarkable but poorly understood regime of transcriptional regulation, whereby individual olfactory neurons each express only one allele of a single member of the large OR gene family. Results We performed a rigorous search for enriched sequence motifs in the largest dataset of OR promoter regions analyzed to date. We combined measures of cross-species conservation with databases of known transcription factor binding sites and ab initio motif-finding algorithms. We found strong enrichment of binding sites for the O/E family of transcription factors and for homeodomain factors, both already known to be involved in the transcriptional control of ORs, but did not identify any novel enriched sequences. We also found that TATA-boxes are present in at least a subset of OR promoters. Conclusions Our rigorous approach provides a template for the analysis of the regulation of large gene families and demonstrates some of the difficulties and pitfalls of such analyses. Although currently available bioinformatics methods cannot detect all transcriptional regulatory elements, our thorough analysis of OR promoters shows that in the case of this gene family, experimental approaches have probably already identified all the binding factors common to large fractions of OR promoters.
Collapse
Affiliation(s)
- Janet M Young
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | |
Collapse
|
27
|
Tsuboi A, Imai T, Kato HK, Matsumoto H, Igarashi KM, Suzuki M, Mori K, Sakano H. Two highly homologous mouse odorant receptors encoded by tandemly-linked MOR29A and MOR29B genes respond differently to phenyl ethers. Eur J Neurosci 2010; 33:205-13. [PMID: 21105914 DOI: 10.1111/j.1460-9568.2010.07495.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Since the discovery of odorant receptors (ORs) in rodents, most ORs have remained orphan receptors. Even for deorphanized ORs in vitro, their in vivo properties are largely unknown. Here, we report odor response profiles of two highly homologous mouse ORs, MOR29A and MOR29B, both in vivo and in vitro. The BAC transgenic mouse was generated, in which olfactory sensory neurons (OSNs) expressing the transgenes MOR29A and MOR29B were differently tagged with IRES-gapECFP and IRES-gapEYFP, respectively. MOR29A- and MOR29B-expressing OSN axons converged on separate but nearby loci on the dorsal surface of the olfactory bulb (OB). Optical imaging of intrinsic signals in the OB identified five different phenyl ethers as candidate ligands for MOR29B. Based on in vitro calcium imaging with the isolated OSNs and luciferase assay with heterologous cells, only guaiacol and vanillin were found to be potent agonists for MOR29A and MOR29B. Because of its accessible glomerular locations in the dorsal OB and defined odor response profiles both in vivo and in vitro, the MOR29A/29B tagging mouse will serve as an excellent tool for studying both odor-signal processing and neural circuitry in the OB.
Collapse
Affiliation(s)
- Akio Tsuboi
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Endo D, Yamamoto Y, Yamaguchi-Yamada M, Nakamuta N, Taniguchi K. Localization of eNOS in the olfactory epithelium of the rat. J Vet Med Sci 2010; 73:423-30. [PMID: 21068516 DOI: 10.1292/jvms.10-0353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide (NO) is a free radical and produced from L-arginine by nitric oxide synthase (NOS). Since NO is recently suggested to be involved in olfactory perception, the expression of eNOS, an isoform of NOS, was examined in the rat olfactory epithelium. The activity of NADPH-diaphorase was also examined as a marker of NOS. In the dorsomedial region of the nasal cavity, intensely positive reactions for NADPH-diaphorase were observed in the entire cytoplasm of sensory cells (olfactory cells). By immunohistochemistry, intensely positive reactions for eNOS were also found in the dorsomedial region of the nasal cavity. These reactions were observed on the free border of the olfactory epithelium. By immunoelectron microscopy, positive reactions for eNOS were found in the cilia of olfactory cells. In addition, in situ hybridization analysis of the olfactory epithelium revealed the expression of eNOS mRNA in the olfactory cells. These results indicate the presence of eNOS in the olfactory cells of the rat, and differential expression of eNOS in the olfactory epithelium depending on the regions of the nasal cavity. In addition, NO produced by eNOS may be involved in olfactory perception in the cilia of olfactory cells.
Collapse
Affiliation(s)
- Daisuke Endo
- Basic Veterinary Science, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | | | | | | | | |
Collapse
|
29
|
Evolving olfactory systems on the fly. Trends Genet 2010; 26:307-16. [PMID: 20537755 DOI: 10.1016/j.tig.2010.04.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 04/20/2010] [Accepted: 04/22/2010] [Indexed: 12/20/2022]
|
30
|
Takeuchi H, Inokuchi K, Aoki M, Suto F, Tsuboi A, Matsuda I, Suzuki M, Aiba A, Serizawa S, Yoshihara Y, Fujisawa H, Sakano H. Sequential arrival and graded secretion of Sema3F by olfactory neuron axons specify map topography at the bulb. Cell 2010; 141:1056-67. [PMID: 20550939 DOI: 10.1016/j.cell.2010.04.041] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 02/12/2010] [Accepted: 03/29/2010] [Indexed: 11/25/2022]
Abstract
In the mouse olfactory system, the anatomical locations of olfactory sensory neurons (OSNs) roughly correlate with their axonal projection sites along the dorsal-ventral (D-V) axis of the olfactory bulb (OB). Here we report that an axon guidance receptor, Neuropilin-2 (Nrp2), and its repulsive ligand, Semaphorin-3F (Sema3F), are expressed by OSNs in a complementary manner that is important for establishing olfactory map topography. Sema3F is secreted by early-arriving axons of OSNs and is deposited at the anterodorsal OB to repel Nrp2-positive axons that arrive later. Sequential arrival of OSN axons as well as the graded and complementary expression of Nrp2 and Sema3F by OSNs help to form the topographic order along the D-V axis.
Collapse
Affiliation(s)
- Haruki Takeuchi
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Meeks JP, Arnson HA, Holy TE. Representation and transformation of sensory information in the mouse accessory olfactory system. Nat Neurosci 2010; 13:723-30. [PMID: 20453853 PMCID: PMC2930753 DOI: 10.1038/nn.2546] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 04/08/2010] [Indexed: 11/20/2022]
Abstract
In mice, nonvolatile social cues are detected and analyzed by the accessory olfactory system (AOS). Here we provide a first view of information processing in the AOS with respect to individual chemical cues. 12 sulfated steroids, recently discovered mouse AOS ligands, caused widespread activity among vomeronasal sensory neurons (VSNs), yet VSN responses clustered into a small number of repeated functional patterns or processing streams. Downstream neurons in the accessory olfactory bulb (AOB) responded to these ligands with enhanced signal/noise compared to VSNs. Although the dendritic connectivity of AOB mitral cells suggests the capacity for broad integration, most sulfated steroid responses were well-modeled by linear excitatory drive from just one VSN processing stream. However, a substantial minority demonstrated multi-stream integration. Most VSN excitation patterns were also observed in the AOB, but excitation by estradiol sulfate processing streams was rare, suggesting AOB circuit organization is specific to the biological relevance of sensed cues.
Collapse
Affiliation(s)
- Julian P Meeks
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | |
Collapse
|
32
|
Takahashi H, Yoshihara SI, Nishizumi H, Tsuboi A. Neuropilin-2 is required for the proper targeting of ventral glomeruli in the mouse olfactory bulb. Mol Cell Neurosci 2010; 44:233-45. [PMID: 20363325 DOI: 10.1016/j.mcn.2010.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 03/19/2010] [Accepted: 03/25/2010] [Indexed: 10/19/2022] Open
Abstract
Recent evidence shows that olfactory sensory neurons expressing a given odorant receptor (OR) are not necessarily confined to one of four zones, rather arranged in an overlapping manner in the olfactory epithelium (OE). In this study, in situ hybridization of OE sections with the OR probes indicated that the OR genes, the mRNAs of which were detected in an array of glomeruli on olfactory bulb (OB) along the anterodorsal/posteroventral (AD/PV) axis, are expressed in subareal zones within the most ventral zone, zone 4, along the dorsomedial/ventrolateral (DM/VL) axis. We also found that Neuropilin-2 (Nrp2) is expressed in a DM-low to VL-high gradient within zone 4 of OE. Furthermore, in Nrp2 mutant mice, we observed multiple glomeruli for zone 4 ORs in OB. These results suggest that the graded expression of Nrp2 in OE is required for the proper targeting of ventral glomeruli along the AD/PV axis in OB.
Collapse
Affiliation(s)
- Hiroo Takahashi
- Laboratory for Molecular Biology of Neural System, Advanced Medical Research Center, Nara Medical University, Nara, Japan
| | | | | | | |
Collapse
|
33
|
Yeshurun Y, Sobel N. An odor is not worth a thousand words: from multidimensional odors to unidimensional odor objects. Annu Rev Psychol 2010; 61:219-41, C1-5. [PMID: 19958179 DOI: 10.1146/annurev.psych.60.110707.163639] [Citation(s) in RCA: 214] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Olfaction is often referred to as a multidimensional sense. It is multidimensional in that approximately 1000 different receptor types, each tuned to particular odor aspects, together contribute to the olfactory percept. In humans, however, this percept is nearly unidimensional. Humans can detect and discriminate countless odorants, but can identify few by name. The one thing humans can and do invariably say about an odor is whether it is pleasant or not. We argue that this hedonic determination is the key function of olfaction. Thus, the boundaries of an odor object are determined by its pleasantness, which--unlike something material and more like an emotion--remains poorly delineated with words.
Collapse
Affiliation(s)
- Yaara Yeshurun
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| | | |
Collapse
|
34
|
Zou DJ, Chesler A, Firestein S. How the olfactory bulb got its glomeruli: a just so story? Nat Rev Neurosci 2009; 10:611-8. [PMID: 19584894 DOI: 10.1038/nrn2666] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The nearly 2,000 glomeruli that cover the surface of the olfactory bulb are so distinctive that they were noted specifically in the earliest of Cajal's catalogues. They have variously been considered a functional unit, an organizational unit and a crucial component of the olfactory coding circuit. Despite their central position in olfactory processing, the development of the glomeruli has only recently begun to be investigated with new and powerful genetic tools. Some unexpected findings have been made that may lead to a new understanding of the processes involved in wiring sensory regions of the brain. It may no longer be sufficient to simply invoke genes, spikes and their interplay in the construction of brain circuits. The story of 'how the olfactory bulb got its glomeruli' may be more complex, and more revealing, than has been supposed.
Collapse
Affiliation(s)
- Dong-Jing Zou
- Department of Biological Sciences, Columbia University, 923 Fairchild Center M.C. 2438 New York, NY 10027, USA
| | | | | |
Collapse
|
35
|
Ortega-Hernandez OD, Kivity S, Shoenfeld Y. Olfaction, psychiatric disorders and autoimmunity: Is there a common genetic association? Autoimmunity 2009; 42:80-8. [DOI: 10.1080/08916930802366140] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
36
|
Sato T, Hirono J, Hamana H, Ishikawa T, Shimizu A, Takashima I, Kajiwara R, Iijima T. Architecture of odor information processing in the olfactory system. Anat Sci Int 2009; 83:195-206. [PMID: 19159347 DOI: 10.1111/j.1447-073x.2007.00215.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Since the discovery of the superfamily of approximately 1000 odorant receptor genes in rodents, the structural simplicity as well as the complexity of the olfactory system have been revealed. The simple aspects include the one neuron-one receptor rule and the exclusive convergence of projections from receptor neurons expressing the same receptors to one or two glomeruli in the olfactory bulb. Odor decoding in the olfactory cortex or higher cortical areas is likely to be a complicated process that depends on the sequence of signal activation and the relative signal intensities of receptors overlapping for similar but different odors. The aim of the present study was to investigate odor information processing both in receptors and in the olfactory cortex. At the receptor level, the similarity and difference in receptor codes between a pair of chiral odorants were examined using the tissue-printing method for sampling all the epithelial zones. In order to dissect odor-driven signal processing in the olfactory cortex by reducing cross-talk with the non-olfactory activities, such as cyclic respiration or other sensory inputs, an in vitro preparation of isolated whole brain with an attached nose was developed, and the methodologies and resulting hypothesis of receptor-sensitivity-dependent hierarchical odor information coding were reviewed.
Collapse
Affiliation(s)
- Takaaki Sato
- Research Institute for Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Amagasaki, Hyogo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Richard M, Sacquet J, Jany M, Schweitzer A, Jourdan F, Andrieux A, Pellier-Monnin V. STOP proteins contribute to the maturation of the olfactory system. Mol Cell Neurosci 2009; 41:120-34. [PMID: 19236915 DOI: 10.1016/j.mcn.2009.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 01/13/2009] [Accepted: 02/06/2009] [Indexed: 01/09/2023] Open
Abstract
Regulation of microtubule dynamics is crucial for axon growth and guidance as well as for the establishment of synaptic connections. STOPs (Stable Tubule Only Polypeptides) are microtubule-associated proteins that regulate microtubule stabilization but are also able to interact with actin or Golgi membranes. Here, we have investigated the involvement of STOPs during the development of the olfactory system. We first describe the spatio-temporal expression patterns of N- and E-STOP, the two neuronal-specific isoforms of STOP. E- and N-STOP are expressed in the axonal compartment of olfactory sensory neurons, but are differentially regulated during development. Interestingly, each neuronal isoform displays a specific gradient distribution within the olfactory nerve layer. Then, we have examined the development of the olfactory system in the absence of STOPs. Olfactory axons display a normal outgrowth and targeting in STOP-null mice, but maturation of the synapses in the glomerular neuropil is altered.
Collapse
Affiliation(s)
- Marion Richard
- Laboratoire Neurosciences Sensorielles, Comportement, Cognition, CNRS-UMR 5020, Université de Lyon, Lyon 1, F-69366, France.
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Gene expression is a fundamentally stochastic process, with randomness in transcription and translation leading to cell-to-cell variations in mRNA and protein levels. This variation appears in organisms ranging from microbes to metazoans, and its characteristics depend both on the biophysical parameters governing gene expression and on gene network structure. Stochastic gene expression has important consequences for cellular function, being beneficial in some contexts and harmful in others. These situations include the stress response, metabolism, development, the cell cycle, circadian rhythms, and aging.
Collapse
Affiliation(s)
- Arjun Raj
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
39
|
Tian H, Ma M. Differential development of odorant receptor expression patterns in the olfactory epithelium: a quantitative analysis in the mouse septal organ. Dev Neurobiol 2008; 68:476-86. [PMID: 18214836 PMCID: PMC2266684 DOI: 10.1002/dneu.20612] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The rodent olfactory epithelium expresses more than 1000 odorant receptors (ORs) with distinct patterns, yet it is unclear how such patterns are established during development. In the current study, we investigated development of the expression patterns of different ORs in the septal organ, a small patch of olfactory epithelium predominantly expressing nine identified ORs. The presumptive septal organ first appears at about embryonic day 16 (E16) and it completely separates from the main olfactory epithelium (MOE) at about postnatal day 7 (P7). Using in situ hybridization, we quantified the densities of the septal organ neurons labeled by specific RNA probes of the nine abundant OR genes from E16 to postnatal 3 months. The results indicate that olfactory sensory neurons (OSNs) expressing different ORs have asynchronous temporal onsets. For instance, MOR256-17 and MOR236-1 cells are present in the septal organ at E16; however, MOR0-2 cells do not appear until P0. In addition, OSNs expressing different ORs show distinct developmental courses and reach their maximum densities at different stages ranging from E16 (e.g. MOR256-17) to 1 month (e.g. MOR256-3 and MOR235-1). Furthermore, early onset does not correlate with high abundance in adult. This study reveals a dynamic composition of the OSNs expressing different ORs in the developing olfactory epithelium.
Collapse
Affiliation(s)
- Huikai Tian
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia 19104, USA
| | | |
Collapse
|
40
|
Biju K, Ronald Marks D, Gerald Mast T, Ann Fadool D. Deletion of voltage-gated channel affects glomerular refinement and odorant receptor expression in the mouse olfactory system. J Comp Neurol 2008; 506:161-79. [PMID: 18022950 PMCID: PMC2650277 DOI: 10.1002/cne.21540] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Olfactory sensory neurons (OSNs) expressing a specific odorant receptor (OR) gene send axonal projections to specific glomeruli, creating a stereotypic olfactory sensory map. Odorant receptor sequence, G-protein cAMP signaling, and axon guidance molecules have been shown to direct axons of OSNs toward central targets in the olfactory bulb (OB). Although the OR sequence may act as one determinant, our objective was to elucidate the extent by which voltage-dependent activity of postsynaptic projection neurons in the OB centrally influences peripheral development and target destination of OSNs. We bred OR-tagged transgenic mice to homozygosity with mice that had a gene-targeted deletion of the Shaker potassium ion channel (Kv1.3) to elucidate how activity modulates synaptic connections that formulate the sensory map. Here we report that the Kv1.3 ion channel, which is predominantly expressed in mitral cells and whose gene-targeted deletion causes a "super-smeller" phenotype, alters synaptic refinement of axonal projections from OSNs expressing P2, M72, and MOR28 ORs. Absence of Kv1.3 voltage-gated activity caused the formation of small, heterogeneous, and supernumerary glomeruli that failed to undergo neural pruning over development. These changes were accompanied by a significant decrease in the number of P2-, M72-, and MOR28-expressing OSNs, which contained an overexpression of OR protein and G-protein G(olf) in the cilia of the olfactory epithelium. These findings suggest that voltage-gated activity of projection neurons is essential to refine primary olfactory projections and that it regulates proper expression of the transduction machinery at the periphery.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- Axons/physiology
- Axons/ultrastructure
- GTP-Binding Proteins/metabolism
- Galactosides/metabolism
- Gene Expression Regulation, Developmental/genetics
- Green Fluorescent Proteins/metabolism
- Indoles/metabolism
- Kv1.3 Potassium Channel/deficiency
- Kv1.3 Potassium Channel/genetics
- Mice
- Mice, Transgenic
- Microscopy, Electron, Scanning/methods
- Mutation
- Neurons, Afferent/cytology
- Neurons, Afferent/metabolism
- Neurons, Afferent/ultrastructure
- Olfactory Marker Protein/metabolism
- Olfactory Pathways/cytology
- Olfactory Pathways/metabolism
- Receptors, Odorant/genetics
- Receptors, Odorant/metabolism
Collapse
Affiliation(s)
- K.C. Biju
- Department of Biological Science, Program in Neuroscience, Biomedical Research Facility, The Florida State University, Tallahassee, Florida 32306
| | - David Ronald Marks
- Department of Biological Science, Program in Neuroscience, Biomedical Research Facility, The Florida State University, Tallahassee, Florida 32306
| | - Thomas Gerald Mast
- Department of Biological Science, Program in Neuroscience, Biomedical Research Facility, The Florida State University, Tallahassee, Florida 32306
| | - Debra Ann Fadool
- Department of Biological Science, Program in Neuroscience, Biomedical Research Facility, The Florida State University, Tallahassee, Florida 32306
- Department of Biological Science, Programs in Neuroscience and Molecular Biophysics, The Florida State University, Tallahassee, Florida 32306
| |
Collapse
|
41
|
Deletion of the core-H region in mice abolishes the expression of three proximal odorant receptor genes in cis. Proc Natl Acad Sci U S A 2007; 104:20067-72. [PMID: 18077433 DOI: 10.1073/pnas.0706544105] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We have previously reported that a 2.1-kb homology (H) sequence, conserved between mouse and human, regulates the odorant receptor (OR) gene MOR28 in transgenic mice. Here, we narrowed down the essential sequences of the H to a core of 124 bp by using a transient expression system in zebrafish embryos. Transgenic experiments in mice demonstrated that the core-H sequence is sufficient to endow expression of the MOR28 minigene. Deletion and mutation analyses of the core-H region revealed two homeodomain sequences to be essential for the H enhancer activity. Targeted deletion of the core-H abolished expression of three proximal OR genes, MOR28, MOR10, and MOR83, in cis, indicating the presence of another locus control region/enhancer in the downstream region, that regulates four distal OR genes in the same MOR28 cluster. In the heterozygous mice, the H(-) phenotype of the mutant allele was not rescued by the wild-type H(+) allele in trans.
Collapse
|
42
|
Nguyen MQ, Zhou Z, Marks CA, Ryba NJP, Belluscio L. Prominent roles for odorant receptor coding sequences in allelic exclusion. Cell 2007; 131:1009-17. [PMID: 18045541 PMCID: PMC2195930 DOI: 10.1016/j.cell.2007.10.050] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Revised: 10/26/2007] [Accepted: 10/30/2007] [Indexed: 01/05/2023]
Abstract
Mammalian odorant receptors (ORs) are crucial for establishing the functional organization of the olfactory system, but the mechanisms controlling their expression remain largely unexplained. Here, we utilized a transgenic approach to explore OR gene regulation. We determined that although olfactory sensory neurons (OSNs) are capable of supporting expression of multiple functional ORs, several levels of control ensure that each neuron normally expresses only a single odorant receptor. Surprisingly, this regulation extends beyond endogenous ORs even preventing expression of transgenes consisting of OR-coding sequences driven by synthetic promoters. Thus, part of the intrinsic feedback system must rely on elements present in the OR-coding sequence. Notably, by expressing the same transgenic ORs precociously in immature neurons, we have overcome this suppression and established a generic method to express any OR in approximately 90% of OSNs. These results provide important insights into the hierarchy of OR gene expression and the vital role of the OR-coding sequence in this regulation.
Collapse
Affiliation(s)
- Minh Q Nguyen
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
43
|
Fuss SH, Omura M, Mombaerts P. Local and cis effects of the H element on expression of odorant receptor genes in mouse. Cell 2007; 130:373-84. [PMID: 17662950 DOI: 10.1016/j.cell.2007.06.023] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2007] [Revised: 03/08/2007] [Accepted: 06/13/2007] [Indexed: 12/18/2022]
Abstract
From the approximately 1,200 odorant receptor (OR) genes in the mouse genome, an olfactory sensory neuron is thought to express only one gene. The mechanisms of OR gene choice are not understood. A 2.1 kilobase region (the H element) adjacent to a cluster of seven OR genes has been proposed as a trans- and pan-enhancer for OR gene expression. Here, we deleted the H element by gene targeting in mice. The deletion abolishes expression of a family of three OR genes proximal to H, and H operates in cis on these genes. Deletion of H has a graded effect on expression of a distal group of four OR genes, commensurate with genomic distance. There is no demonstrable effect on expression of OR genes located outside the cluster. Our findings are not consistent with the hypothesis of H as an essential trans-acting enhancer for genome-wide regulation of OR gene expression.
Collapse
Affiliation(s)
- Stefan H Fuss
- The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | | | | |
Collapse
|
44
|
Abstract
Systematic mapping studies involving 365 odorant chemicals have shown that glomerular responses in the rat olfactory bulb are organized spatially in patterns that are related to the chemistry of the odorant stimuli. This organization involves the spatial clustering of principal responses to numerous odorants that share key aspects of chemistry such as functional groups, hydrocarbon structural elements, and/or overall molecular properties related to water solubility. In several of the clusters, responses shift progressively in position according to odorant carbon chain length. These response domains appear to be constructed from orderly projections of sensory neurons in the olfactory epithelium and may also involve chromatography across the nasal mucosa. The spatial clustering of glomerular responses may serve to "tune" the principal responses of bulbar projection neurons by way of inhibitory interneuronal networks, allowing the projection neurons to respond to a narrower range of stimuli than their associated sensory neurons. When glomerular activity patterns are viewed relative to the overall level of glomerular activation, the patterns accurately predict the perception of odor quality, thereby supporting the notion that spatial patterns of activity are the key factors underlying that aspect of the olfactory code. A critical analysis suggests that alternative coding mechanisms for odor quality, such as those based on temporal patterns of responses, enjoy little experimental support.
Collapse
Affiliation(s)
- Brett A Johnson
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4550, USA.
| | | |
Collapse
|
45
|
Serizawa S, Miyamichi K, Takeuchi H, Yamagishi Y, Suzuki M, Sakano H. A Neuronal Identity Code for the Odorant Receptor-Specific and Activity-Dependent Axon Sorting. Cell 2006; 127:1057-69. [PMID: 17129788 DOI: 10.1016/j.cell.2006.10.031] [Citation(s) in RCA: 261] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Revised: 09/14/2006] [Accepted: 10/18/2006] [Indexed: 10/23/2022]
Abstract
In the mouse, olfactory sensory neurons (OSNs) expressing the same odorant receptor (OR) converge their axons to a specific set of glomeruli in the olfactory bulb. To study how OR-instructed axonal fasciculation is controlled, we searched for genes whose expression profiles are correlated with the expressed ORs. Using the transgenic mouse in which the majority of OSNs express a particular OR, we identified such genes coding for the homophilic adhesive molecules Kirrel2/Kirrel3 and repulsive molecules ephrin-A5/EphA5. In the CNGA2 knockout mouse, where the odor-evoked cation influx is disrupted, Kirrel2 and EphA5 were downregulated, while Kirrel3 and ephrin-A5 were upregulated, indicating that these genes are transcribed in an activity-dependent manner. Mosaic analysis demonstrated that gain of function of these genes generates duplicated glomeruli. We propose that a specific set of adhesive/repulsive molecules, whose expression levels are determined by OR molecules, regulate the axonal fasciculation of OSNs during the process of glomerular map formation.
Collapse
Affiliation(s)
- Shou Serizawa
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0032, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Wagner S, Gresser AL, Torello AT, Dulac C. A multireceptor genetic approach uncovers an ordered integration of VNO sensory inputs in the accessory olfactory bulb. Neuron 2006; 50:697-709. [PMID: 16731509 DOI: 10.1016/j.neuron.2006.04.033] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 04/17/2006] [Accepted: 04/26/2006] [Indexed: 11/29/2022]
Abstract
Pheromone detection by the vomeronasal organ (VNO) is thought to rely on activation of specific receptors from the V1R and V2R gene families, but the central representation of pheromone receptor activation remains poorly understood. We generated transgenic mouse lines in which projections from multiple populations of VNO neurons, each expressing a distinct V1R, are differentially labeled with fluorescent proteins. This approach revealed that inputs from neurons expressing closely related V1Rs intermingle within shared, spatially conserved domains of the accessory olfactory bulb (AOB). Mitral cell-glomerular connectivity was examined by injecting intracellular dyes into AOB mitral cells and monitoring dendritic contacts with genetically labeled glomeruli. We show that individual mitral cells extend dendrites to glomeruli associated with different, but likely closely related, V1Rs. This organization differs from the labeled line of OR signaling in the main olfactory system and suggests that integration of information may already occur at the level of the AOB.
Collapse
Affiliation(s)
- Shlomo Wagner
- Department of Neurobiology, Hebrew University, Jerusalem 91904, Israel.
| | | | | | | |
Collapse
|
47
|
Tsuboi A, Miyazaki T, Imai T, Sakano H. Olfactory sensory neurons expressing class I odorant receptors converge their axons on an antero-dorsal domain of the olfactory bulb in the mouse. Eur J Neurosci 2006; 23:1436-44. [PMID: 16553607 DOI: 10.1111/j.1460-9568.2006.04675.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vertebrate odorant receptor (OR) genes are divided phylogenetically into two distinct classes: the fish-like class I and the terrestrial-specific class II. In the present study, we systematically analysed mouse class I OR genes (42 subfamilies) to elucidate the expression profiles in the olfactory epithelium (OE) and the projection sites of their olfactory sensory neurons (OSNs) in the olfactory bulb (OB). In situ hybridization (ISH) revealed that most class I OR genes (36 subfamilies) were expressed in the dorso-medial zone (zone 1) of the OE. Furthermore, there appeared to be no significant differences in the distributions of OSNs expressing class I genes within zone 1. These results indicate that there is a clear boundary between zone 1 and non-zone 1 areas in the OE. Some class I ORs are known to possess ligand specificity for aliphatic acids, aldehydes and alcohols. Our ISH analysis has revealed that OSNs expressing the class I ORs in zone 1 tend to converge their axons on a cluster of glomeruli in an antero-dorsal domain that is assumed to be involved in responses to the aliphatic compounds on the OB.
Collapse
Affiliation(s)
- Akio Tsuboi
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Japan.
| | | | | | | |
Collapse
|
48
|
Mori K, Takahashi YK, Igarashi KM, Yamaguchi M. Maps of odorant molecular features in the Mammalian olfactory bulb. Physiol Rev 2006; 86:409-33. [PMID: 16601265 DOI: 10.1152/physrev.00021.2005] [Citation(s) in RCA: 255] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The olfactory bulb (OB) is the first relay station of the central olfactory system in the mammalian brain and contains a few thousand glomeruli on its surface. Because individual glomeruli represent a single odorant receptor, the glomerular sheet of the OB forms odorant receptor maps. This review summarizes the emerging view of the spatial organization of the odorant receptor maps. Recent studies suggest that individual odorant receptors are molecular-feature detecting units, and so are individual glomeruli in the OB. How are the molecular-feature detecting units spatially arranged in the glomerular sheet? To characterize the molecular-feature specificity of an individual glomerulus, it is necessary to determine the molecular receptive range (MRR) of the glomerulus and to compare the molecular structure of odorants within the MRR. Studies of the MRR mapping show that 1) individual glomeruli typically respond to a range of odorants that share a specific combination of molecular features, 2) each glomerulus appears to be unique in its MRR property, and 3) glomeruli with similar MRR properties gather together in proximity and form molecular-feature clusters. The molecular-feature clusters are located at stereotypical positions in the OB and might be part of the neural representation of basic odor quality. Detailed studies suggest that the glomerular sheet represents the characteristic molecular features in a systematic, gradual, and multidimensional fashion. The molecular-feature maps provide a basis for understanding how the olfactory cortex reads the odor maps of the OB.
Collapse
Affiliation(s)
- Kensaku Mori
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
| | | | | | | |
Collapse
|
49
|
Hoppe R, Breer H, Strotmann J. Promoter motifs of olfactory receptor genes expressed in distinct topographic patterns. Genomics 2006; 87:711-23. [PMID: 16600568 DOI: 10.1016/j.ygeno.2006.02.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2005] [Revised: 11/07/2005] [Accepted: 02/09/2006] [Indexed: 11/25/2022]
Abstract
Novel olfactory receptor-encoding genes that are expressed in olfactory sensory neurons arranged in a clustered pattern in the nasal epithelium, typical of the mOR262 (approved gene symbol Olfr) family, were identified. The genes share sequence motifs upstream of their transcription start sites that are highly related to those previously identified as characteristic of the mOR262 genes, suggesting that these regulatory elements may contribute to governing their unique expression pattern. Promoter analyses of genes encoding class I receptors that are expressed in the dorsal region of the epithelium revealed a different, but again common set of sequence motifs. A prominent feature of the class I gene promoters are multiple O/E-like binding sites, and O/E-type transcription factors that bind to the putative promoter region of class I OR genes were in fact identified. The findings support the concept that common elements in the promoter region of these OR genes may determine their congenic expression pattern in the epithelium.
Collapse
Affiliation(s)
- Reiner Hoppe
- Institute of Physiology, University of Hohenheim, Garbenstrasse 30, 70593 Stuttgart, Germany
| | | | | |
Collapse
|
50
|
Abstract
The act of sniffing increases the air velocity and changes the duration of airflow in the nose. It is not yet clear how these changes interact with the intrinsic timing within the olfactory bulb, but this is a matter of current research activity. An action of sniffing in generating a high velocity that alters the sorption of odorants onto the lining of the nasal cavity is expected from the established work on odorant properties and sorption in the frog nose. Recent work indicates that the receptor properties in the olfactory epithelium and olfactory bulb are correlated with the receptor gene expression zones. The responses in both the epithelium and the olfactory bulb are predictable to a considerable extent by the hydrophobicity of odorants. Furthermore, receptor expression in both rodent and salamander nose interacts with the shapes of the nasal cavity to place the receptor sensitivity to odorants in optimal places according to the aerodynamic properties of the nose.
Collapse
Affiliation(s)
- John W Scott
- Department of Cell Biology, Emory University, 405N Whitehead Biomedical Research Center, 615 Michael Street, Atlanta, GA 30322, USA.
| |
Collapse
|