1
|
Stark R. The olfactory bulb: A neuroendocrine spotlight on feeding and metabolism. J Neuroendocrinol 2024; 36:e13382. [PMID: 38468186 DOI: 10.1111/jne.13382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/13/2024]
Abstract
Olfaction is the most ancient sense and is needed for food-seeking, danger protection, mating and survival. It is often the first sensory modality to perceive changes in the external environment, before sight, taste or sound. Odour molecules activate olfactory sensory neurons that reside on the olfactory epithelium in the nasal cavity, which transmits this odour-specific information to the olfactory bulb (OB), where it is relayed to higher brain regions involved in olfactory perception and behaviour. Besides odour processing, recent studies suggest that the OB extends its function into the regulation of food intake and energy balance. Furthermore, numerous hormone receptors associated with appetite and metabolism are expressed within the OB, suggesting a neuroendocrine role outside the hypothalamus. Olfactory cues are important to promote food preparatory behaviours and consumption, such as enhancing appetite and salivation. In addition, altered metabolism or energy state (fasting, satiety and overnutrition) can change olfactory processing and perception. Similarly, various animal models and human pathologies indicate a strong link between olfactory impairment and metabolic dysfunction. Therefore, understanding the nature of this reciprocal relationship is critical to understand how olfactory or metabolic disorders arise. This present review elaborates on the connection between olfaction, feeding behaviour and metabolism and will shed light on the neuroendocrine role of the OB as an interface between the external and internal environments. Elucidating the specific mechanisms by which olfactory signals are integrated and translated into metabolic responses holds promise for the development of targeted therapeutic strategies and interventions aimed at modulating appetite and promoting metabolic health.
Collapse
Affiliation(s)
- Romana Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
2
|
Raja R, Dumontier E, Phen A, Cloutier JF. Insertion of a neomycin selection cassette in the Amigo1 locus alters gene expression in the olfactory epithelium leading to region-specific defects in olfactory receptor neuron development. Genesis 2024; 62:e23594. [PMID: 38590146 DOI: 10.1002/dvg.23594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/10/2024]
Abstract
During development of the nervous system, neurons connect to one another in a precisely organized manner. Sensory systems provide a good example of this organization, whereby the composition of the outside world is represented in the brain by neuronal maps. Establishing correct patterns of neural circuitry is crucial, as inaccurate map formation can lead to severe disruptions in sensory processing. In rodents, olfactory stimuli modulate a wide variety of behaviors essential for survival. The formation of the olfactory glomerular map is dependent on molecular cues that guide olfactory receptor neuron axons to broad regions of the olfactory bulb and on cell adhesion molecules that promote axonal sorting into specific synaptic units in this structure. Here, we demonstrate that the cell adhesion molecule Amigo1 is expressed in a subpopulation of olfactory receptor neurons, and we investigate its role in the precise targeting of olfactory receptor neuron axons to the olfactory bulb using a genetic loss-of-function approach in mice. While ablation of Amigo1 did not lead to alterations in olfactory sensory neuron axonal targeting, our experiments revealed that the presence of a neomycin resistance selection cassette in the Amigo1 locus can lead to off-target effects that are not due to loss of Amigo1 expression, including unexpected altered gene expression in olfactory receptor neurons and reduced glomerular size in the ventral region of the olfactory bulb. Our results demonstrate that insertion of a neomycin selection cassette into the mouse genome can have specific deleterious effects on the development of the olfactory system and highlight the importance of removing antibiotic resistance cassettes from genetic loss-of-function mouse models when studying olfactory system development.
Collapse
Affiliation(s)
- Reesha Raja
- The Neuro (Montreal Neurological Institute-Hospital), Montréal, Québec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - Emilie Dumontier
- The Neuro (Montreal Neurological Institute-Hospital), Montréal, Québec, Canada
| | - Alina Phen
- The Neuro (Montreal Neurological Institute-Hospital), Montréal, Québec, Canada
| | - Jean-François Cloutier
- The Neuro (Montreal Neurological Institute-Hospital), Montréal, Québec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| |
Collapse
|
3
|
Hirota J. Molecular mechanisms of differentiation and class choice of olfactory sensory neurons. Genesis 2024; 62:e23587. [PMID: 38454646 DOI: 10.1002/dvg.23587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/10/2024] [Accepted: 01/29/2024] [Indexed: 03/09/2024]
Abstract
The sense of smell is intricately linked to essential animal behaviors necessary for individual survival and species preservation. During vertebrate evolution, odorant receptors (ORs), responsible for detecting odor molecules, have evolved to adapt to changing environments, transitioning from aquatic to terrestrial habitats and accommodating increasing complex chemical environments. These evolutionary pressures have given rise to the largest gene family in vertebrate genomes. Vertebrate ORs are phylogenetically divided into two major classes; class I and class II. Class I OR genes, initially identified in fish and frog, have persisted across vertebrate species. On the other hand, class II OR genes are unique to terrestrial animals, accounting for ~90% of mammalian OR genes. In mice, each olfactory sensory neuron (OSN) expresses a single functional allele of a single OR gene from either the class I or class II OR repertoire. This one neuron-one receptor rule is established through two sequential steps: specification of OR class and subsequent exclusive OR expression from the corresponding OR class. Consequently, OSNs acquire diverse neuronal identities during the process of OSN differentiation, enabling animals to detect a wide array of odor molecules. This review provides an overview of the OSN differentiation process through which OSN diversity is achieved, primarily using the mouse as a model animal.
Collapse
Affiliation(s)
- Junji Hirota
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- Center for Integrative Biosciences, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
4
|
Fang A, Yu CR. Activity-dependent formation of the topographic map and the critical period in the development of mammalian olfactory system. Genesis 2024; 62:e23586. [PMID: 38593162 PMCID: PMC11003738 DOI: 10.1002/dvg.23586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 04/11/2024]
Abstract
Neural activity influences every aspect of nervous system development. In olfactory systems, sensory neurons expressing the same odorant receptor project their axons to stereotypically positioned glomeruli, forming a spatial map of odorant receptors in the olfactory bulb. As individual odors activate unique combinations of glomeruli, this map forms the basis for encoding olfactory information. The establishment of this stereotypical olfactory map requires coordinated regulation of axon guidance molecules instructed by spontaneous activity. Recent studies show that sensory experiences also modify innervation patterns in the olfactory bulb, especially during a critical period of the olfactory system development. This review examines evidence in the field to suggest potential mechanisms by which various aspects of neural activity regulate axon targeting. We also discuss the precise functions served by neural plasticity during the critical period.
Collapse
Affiliation(s)
- Ai Fang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| |
Collapse
|
5
|
Toh H, Yang C, Formenti G, Raja K, Yan L, Tracey A, Chow W, Howe K, Bergeron LA, Zhang G, Haase B, Mountcastle J, Fedrigo O, Fogg J, Kirilenko B, Munegowda C, Hiller M, Jain A, Kihara D, Rhie A, Phillippy AM, Swanson SA, Jiang P, Clegg DO, Jarvis ED, Thomson JA, Stewart R, Chaisson MJP, Bukhman YV. A haplotype-resolved genome assembly of the Nile rat facilitates exploration of the genetic basis of diabetes. BMC Biol 2022; 20:245. [DOI: 10.1186/s12915-022-01427-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
The Nile rat (Avicanthis niloticus) is an important animal model because of its robust diurnal rhythm, a cone-rich retina, and a propensity to develop diet-induced diabetes without chemical or genetic modifications. A closer similarity to humans in these aspects, compared to the widely used Mus musculus and Rattus norvegicus models, holds the promise of better translation of research findings to the clinic.
Results
We report a 2.5 Gb, chromosome-level reference genome assembly with fully resolved parental haplotypes, generated with the Vertebrate Genomes Project (VGP). The assembly is highly contiguous, with contig N50 of 11.1 Mb, scaffold N50 of 83 Mb, and 95.2% of the sequence assigned to chromosomes. We used a novel workflow to identify 3613 segmental duplications and quantify duplicated genes. Comparative analyses revealed unique genomic features of the Nile rat, including some that affect genes associated with type 2 diabetes and metabolic dysfunctions. We discuss 14 genes that are heterozygous in the Nile rat or highly diverged from the house mouse.
Conclusions
Our findings reflect the exceptional level of genomic resolution present in this assembly, which will greatly expand the potential of the Nile rat as a model organism.
Collapse
|
6
|
Voortman L, Anderson C, Urban E, Yuan L, Tran S, Neuhaus-Follini A, Derrick J, Gregor T, Johnston RJ. Temporally dynamic antagonism between transcription and chromatin compaction controls stochastic photoreceptor specification in flies. Dev Cell 2022; 57:1817-1832.e5. [PMID: 35835116 PMCID: PMC9378680 DOI: 10.1016/j.devcel.2022.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 05/09/2022] [Accepted: 06/20/2022] [Indexed: 01/12/2023]
Abstract
Stochastic mechanisms diversify cell fates during development. How cells randomly choose between two or more fates remains poorly understood. In the Drosophila eye, the random mosaic of two R7 photoreceptor subtypes is determined by expression of the transcription factor Spineless (Ss). We investigated how cis-regulatory elements and trans factors regulate nascent transcriptional activity and chromatin compaction at the ss gene locus during R7 development. The ss locus is in a compact state in undifferentiated cells. An early enhancer drives transcription in all R7 precursors, and the locus opens. In differentiating cells, transcription ceases and the ss locus stochastically remains open or compacts. In SsON R7s, ss is open and competent for activation by a late enhancer, whereas in SsOFF R7s, ss is compact, and repression prevents expression. Our results suggest that a temporally dynamic antagonism, in which transcription drives large-scale decompaction and then compaction represses transcription, controls stochastic fate specification.
Collapse
Affiliation(s)
- Lukas Voortman
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Caitlin Anderson
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Elizabeth Urban
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Luorongxin Yuan
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sang Tran
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Josh Derrick
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Thomas Gregor
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Joseph Henry Laboratories of Physics, the Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Developmental and Stem Cell Biology, UMR3738, Institut Pasteur, 75015 Paris, France
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
7
|
Ruiz Tejada Segura ML, Abou Moussa E, Garabello E, Nakahara TS, Makhlouf M, Mathew LS, Wang L, Valle F, Huang SSY, Mainland JD, Caselle M, Osella M, Lorenz S, Reisert J, Logan DW, Malnic B, Scialdone A, Saraiva LR. A 3D transcriptomics atlas of the mouse nose sheds light on the anatomical logic of smell. Cell Rep 2022; 38:110547. [PMID: 35320714 PMCID: PMC8995392 DOI: 10.1016/j.celrep.2022.110547] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/26/2022] [Accepted: 03/01/2022] [Indexed: 12/26/2022] Open
Abstract
The sense of smell helps us navigate the environment, but its molecular architecture and underlying logic remain understudied. The spatial location of odorant receptor genes (Olfrs) in the nose is thought to be independent of the structural diversity of the odorants they detect. Using spatial transcriptomics, we create a genome-wide 3D atlas of the mouse olfactory mucosa (OM). Topographic maps of genes differentially expressed in space reveal that both Olfrs and non-Olfrs are distributed in a continuous and overlapping fashion over at least five broad zones in the OM. The spatial locations of Olfrs correlate with the mucus solubility of the odorants they recognize, providing direct evidence for the chromatographic theory of olfaction. This resource resolves the molecular architecture of the mouse OM and will inform future studies on mechanisms underlying Olfr gene choice, axonal pathfinding, patterning of the nervous system, and basic logic for the peripheral representation of smell.
Collapse
Affiliation(s)
- Mayra L Ruiz Tejada Segura
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Feodor-Lynen-Strasse 21, 81377 München, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | | | - Elisa Garabello
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Feodor-Lynen-Strasse 21, 81377 München, Germany; Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy; Department of Civil and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Thiago S Nakahara
- Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | | | | | - Li Wang
- Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Filippo Valle
- Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy
| | | | - Joel D Mainland
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michele Caselle
- Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy
| | - Matteo Osella
- Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy
| | - Stephan Lorenz
- Sidra Medicine, P.O. Box 26999, Doha, Qatar; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Johannes Reisert
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Darren W Logan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Bettina Malnic
- Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Feodor-Lynen-Strasse 21, 81377 München, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | - Luis R Saraiva
- Sidra Medicine, P.O. Box 26999, Doha, Qatar; Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA; College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar.
| |
Collapse
|
8
|
Odorant-receptor-mediated regulation of chemosensory gene expression in the malaria mosquito Anopheles gambiae. Cell Rep 2022; 38:110494. [PMID: 35263579 PMCID: PMC8957105 DOI: 10.1016/j.celrep.2022.110494] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/03/2022] [Accepted: 02/14/2022] [Indexed: 01/07/2023] Open
Abstract
Mosquitoes locate and approach humans based on the activity of odorant receptors (ORs) expressed on olfactory receptor neurons (ORNs). Olfactogenetic experiments in Anopheles gambiae mosquitoes revealed that the ectopic expression of an AgOR (AgOR2) in ORNs dampened the activity of the expressing neuron. This contrasts with studies in Drosophila melanogaster in which the ectopic expression of non-native ORs in ORNs confers ectopic neuronal responses without interfering with native olfactory physiology. RNA-seq analyses comparing wild-type antennae to those ectopically expressing AgOR2 in ORNs indicated that nearly all AgOR transcripts were significantly downregulated (except for AgOR2). Additional experiments suggest that AgOR2 protein rather than mRNA mediates this downregulation. Using in situ hybridization, we find that AgOR gene choice is active into adulthood and that AgOR2 expression inhibits AgORs from turning on at this late stage. Our study shows that the ORNs of Anopheles mosquitoes (in contrast to Drosophila) are sensitive to a currently unexplored mechanism of AgOR regulation. Maguire et al. discover that the ectopic expression of an olfactory receptor can downregulate the transcription of endogenous odorant receptors in mosquito olfactory neurons. The onset of mosquito odorant-receptor expression by an olfactory neuron continues into adult stages, and is particularly sensitive to exogenous olfactory reception expression.
Collapse
|
9
|
Coppola DM. The sorption/chromatography hypothesis of olfactory discrimination: The rise, fall, and rebirth of a Phoenix. Bioessays 2022; 44:e2100263. [PMID: 34984707 DOI: 10.1002/bies.202100263] [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: 11/07/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/09/2022]
Abstract
Herein, I discuss the enduring mystery of the receptor layout in the vertebrate olfactory system. Since the awarding of the 2004 Nobel Prize to Axel and Buck for their discovery of the gene family that encodes olfactory receptors, our field has enjoyed a golden era. Despite this Renaissance, an answer to one of the most fundamental questions for any sensory system-what is the anatomical logic of its receptor array?-eludes us, still, for olfaction! Indeed, the only widely debated hypothesis, finding its origins in the musing of another Nobel laureate Sir Edgar Adrian, has it that the vertebrate nose organizes its receptors according to the "sorptive" properties of their ligands. This idea, known as the "sorption" or "chromatography" hypothesis, enjoys considerable support despite being controversial. Here, I review the history of the hypothesis-its rises and falls-and discuss the latest data and future prospects for this perennial idea whose history I liken to the mythical Phoenix.
Collapse
Affiliation(s)
- David M Coppola
- Department of Biology, Randolph-Macon College, Ashland, Virginia, 23005, USA
| |
Collapse
|
10
|
Aoki M, Gamayun I, Wyatt A, Grünewald R, Simon-Thomas M, Philipp SE, Hummel O, Wagenpfeil S, Kattler K, Gasparoni G, Walter J, Qiao S, Grattan DR, Boehm U. Prolactin-sensitive olfactory sensory neurons regulate male preference in female mice by modulating responses to chemosensory cues. SCIENCE ADVANCES 2021; 7:eabg4074. [PMID: 34623921 PMCID: PMC8500514 DOI: 10.1126/sciadv.abg4074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 08/19/2021] [Indexed: 06/10/2023]
Abstract
Chemosensory cues detected in the nose need to be integrated with the hormonal status to trigger appropriate behaviors, but the neural circuits linking the olfactory and the endocrine system are insufficiently understood. Here, we characterize olfactory sensory neurons in the murine nose that respond to the pituitary hormone prolactin. Deletion of prolactin receptor in these cells results in impaired detection of social odors and blunts male preference in females. The prolactin-responsive olfactory sensory neurons exhibit a distinctive projection pattern to the brain that is similar across different individuals and express a limited subset of chemosensory receptors. Prolactin modulates the responses within these neurons to discrete chemosensory cues contained in male urine, providing a mechanism by which the hormonal status can be directly linked with distinct olfactory cues to generate appropriate behavioral responses.
Collapse
Affiliation(s)
- Mari Aoki
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Igor Gamayun
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Ramona Grünewald
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Martin Simon-Thomas
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Stephan E. Philipp
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Oliver Hummel
- Faculty of Computer Science, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Stefan Wagenpfeil
- Institute for Medical Biometry, Epidemiology and Medical Informatics, Saarland University School of Medicine, Homburg, Germany
| | - Kathrin Kattler
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Gilles Gasparoni
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Jörn Walter
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Sen Qiao
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - David R. Grattan
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Ulrich Boehm
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| |
Collapse
|
11
|
Zapiec B, Mombaerts P. The Zonal Organization of Odorant Receptor Gene Choice in the Main Olfactory Epithelium of the Mouse. Cell Rep 2021; 30:4220-4234.e5. [PMID: 32209480 DOI: 10.1016/j.celrep.2020.02.110] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/16/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
A mature olfactory sensory neuron (OSN) of the main olfactory epithelium (MOE) typically expresses one allele of one odorant receptor (OR) gene. It is widely thought that the great majority of the 1,141 intact mouse OR genes are expressed in one of four MOE zones (or bands or stripes), which are largely non-overlapping. Here, we develop a multiplex method to map, in 3D and MOE-wide, the expression areas of multiple OR genes in individual, non-genetically modified mice by three-color fluorescence in situ hybridization, semi-automated image segmentation, and 3D reconstruction. We classify the expression areas of 68 OR genes into 9 zones. These zones are highly overlapping and strikingly complex when viewed in 3D reconstructions. There could well be more zones. We propose that zones reflect distinct OSN types that are each restricted in their choice to a subset of the OR gene repertoire.
Collapse
Affiliation(s)
- Bolek Zapiec
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, 60438 Frankfurt, Germany
| | - Peter Mombaerts
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, 60438 Frankfurt, Germany.
| |
Collapse
|
12
|
Son G, Yoo SJ, Kang S, Rasheed A, Jung DH, Park H, Cho B, Steinbusch HWM, Chang KA, Suh YH, Moon C. Region-specific amyloid-β accumulation in the olfactory system influences olfactory sensory neuronal dysfunction in 5xFAD mice. ALZHEIMERS RESEARCH & THERAPY 2021; 13:4. [PMID: 33397474 PMCID: PMC7784287 DOI: 10.1186/s13195-020-00730-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/23/2020] [Indexed: 01/10/2023]
Abstract
Background Hyposmia in Alzheimer’s disease (AD) is a typical early symptom according to numerous previous clinical studies. Although amyloid-β (Aβ), which is one of the toxic factors upregulated early in AD, has been identified in many studies, even in the peripheral areas of the olfactory system, the pathology involving olfactory sensory neurons (OSNs) remains poorly understood. Methods Here, we focused on peripheral olfactory sensory neurons (OSNs) and delved deeper into the direct relationship between pathophysiological and behavioral results using odorants. We also confirmed histologically the pathological changes in 3-month-old 5xFAD mouse models, which recapitulates AD pathology. We introduced a numeric scale histologically to compare physiological phenomenon and local tissue lesions regardless of the anatomical plane. Results We observed the odorant group that the 5xFAD mice showed reduced responses to odorants. These also did not physiologically activate OSNs that propagate their axons to the ventral olfactory bulb. Interestingly, the amount of accumulated amyloid-β (Aβ) was high in the OSNs located in the olfactory epithelial ectoturbinate and the ventral olfactory bulb glomeruli. We also observed irreversible damage to the ectoturbinate of the olfactory epithelium by measuring the impaired neuronal turnover ratio from the basal cells to the matured OSNs. Conclusions Our results showed that partial and asymmetrical accumulation of Aβ coincided with physiologically and structurally damaged areas in the peripheral olfactory system, which evoked hyporeactivity to some odorants. Taken together, partial olfactory dysfunction closely associated with peripheral OSN’s loss could be a leading cause of AD-related hyposmia, a characteristic of early AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-020-00730-2.
Collapse
Affiliation(s)
- Gowoon Son
- Department of Brain & Cognitive Sciences, Graduate School, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Seung-Jun Yoo
- Department of Brain & Cognitive Sciences, Graduate School, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.,Convergence Research Advanced Centre for Olfaction, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.,Max Planck Research Unit for Neurogenetics, Frankfurt, Germany
| | - Shinwoo Kang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Ameer Rasheed
- Department of Brain & Cognitive Sciences, Graduate School, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Da Hae Jung
- Department of Brain & Cognitive Sciences, Graduate School, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Hyunjun Park
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Bongki Cho
- Department of Brain & Cognitive Sciences, Graduate School, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.,Convergence Research Advanced Centre for Olfaction, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Harry W M Steinbusch
- Department of Brain & Cognitive Sciences, Graduate School, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Keun-A Chang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Yoo-Hun Suh
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Cheil Moon
- Department of Brain & Cognitive Sciences, Graduate School, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea. .,Convergence Research Advanced Centre for Olfaction, Daegu Gyeungbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea. .,Korea Brain Research Institute, Daegu, Republic of Korea.
| |
Collapse
|
13
|
Soelter J, Schumacher J, Spors H, Schmuker M. Computational exploration of molecular receptive fields in the olfactory bulb reveals a glomerulus-centric chemical map. Sci Rep 2020; 10:77. [PMID: 31919393 PMCID: PMC6952415 DOI: 10.1038/s41598-019-56863-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 09/24/2019] [Indexed: 01/13/2023] Open
Abstract
Progress in olfactory research is currently hampered by incomplete knowledge about chemical receptive ranges of primary receptors. Moreover, the chemical logic underlying the arrangement of computational units in the olfactory bulb has still not been resolved. We undertook a large-scale approach at characterising molecular receptive ranges (MRRs) of glomeruli in the dorsal olfactory bulb (dOB) innervated by the MOR18-2 olfactory receptor, also known as Olfr78, with human ortholog OR51E2. Guided by an iterative approach that combined biological screening and machine learning, we selected 214 odorants to characterise the response of MOR18-2 and its neighbouring glomeruli. We found that a combination of conventional physico-chemical and vibrational molecular descriptors performed best in predicting glomerular responses using nonlinear Support-Vector Regression. We also discovered several previously unknown odorants activating MOR18-2 glomeruli, and obtained detailed MRRs of MOR18-2 glomeruli and their neighbours. Our results confirm earlier findings that demonstrated tunotopy, that is, glomeruli with similar tuning curves tend to be located in spatial proximity in the dOB. In addition, our results indicate chemotopy, that is, a preference for glomeruli with similar physico-chemical MRR descriptions being located in spatial proximity. Together, these findings suggest the existence of a partial chemical map underlying glomerular arrangement in the dOB. Our methodology that combines machine learning and physiological measurements lights the way towards future high-throughput studies to deorphanise and characterise structure-activity relationships in olfaction.
Collapse
Affiliation(s)
- Jan Soelter
- Neuroinformatics & Theoretical Neuroscience, Freie Universität Berlin, Königin-Luise-Str. 1-3, 14195, Berlin, Germany
| | - Jan Schumacher
- Max-Planck-Institute for Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt/Main, Germany
| | - Hartwig Spors
- Max-Planck-Institute for Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt/Main, Germany
- Department of Neuropediatrics, Max-Liebig-University, Giessen, Germany
| | - Michael Schmuker
- Neuroinformatics & Theoretical Neuroscience, Freie Universität Berlin, Königin-Luise-Str. 1-3, 14195, Berlin, Germany.
- Biocomputation Group, University of Hertfordshire, Hatfield, AL10 9AB, United Kingdom.
| |
Collapse
|
14
|
Coppola DM, Fitzwater E, Rygg AD, Craven BA. Tests of the chromatographic theory of olfaction with highly soluble odors: a combined electro-olfactogram and computational fluid dynamics study in the mouse. Biol Open 2019; 8:bio.047217. [PMID: 31649069 PMCID: PMC6826284 DOI: 10.1242/bio.047217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The idea that the vertebrate nasal cavity operates like a gas chromatograph to separate and discriminate odors, referred to herein as the ‘chromatographic theory’ (CT), has a long and interesting history. Though the last decade has seen renewed interest in the notion, its validity remains in question. Here we examine a necessary condition of the theory: a correlation between nasal odor deposition patterns based on mucus solubility and the distribution of olfactory sensory neuron odotypes. Our recent work in the mouse failed to find such a relationship even across large sorption gradients within the olfactory epithelium (OE). However, these studies did not test extremely soluble odorants or low odor concentrations, factors that could explain our inability to find supporting evidence for the CT. The current study combined computational fluid dynamics (CFD) simulations of odor sorption patterns and electro-olfactogram (EOG) measurements of olfactory sensory neuron responses. The odorants tested were at the extremes of mucus solubility and at a range of concentrations. Results showed no relationship between local odor sorption patterns and EOG response maps. Together, results again failed to support a necessary condition of the CT casting further doubt on the viability of this classical odor coding mechanism. Summary: This paper casts doubt on the classical chromatographic theory of olfaction, showing there is no correlation between olfactory receptor spatial layout and odor solubility patterns, a necessary condition of the theory.
Collapse
Affiliation(s)
- David M Coppola
- Department of Biology, Randolph-Macon College, Ashland, VA 23005, USA
| | - Emily Fitzwater
- Department of Biology, Randolph-Macon College, Ashland, VA 23005, USA
| | - Alex D Rygg
- Department of Ecology and Evolutionary Biology, UCLA, Los Angeles, CA 90095, USA
| | - Brent A Craven
- Department of Mechanical and Nuclear Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
15
|
Liberia T, Martin-Lopez E, Meller SJ, Greer CA. Sequential Maturation of Olfactory Sensory Neurons in the Mature Olfactory Epithelium. eNeuro 2019; 6:ENEURO.0266-19.2019. [PMID: 31554664 PMCID: PMC6795559 DOI: 10.1523/eneuro.0266-19.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/10/2019] [Accepted: 09/18/2019] [Indexed: 12/20/2022] Open
Abstract
The formation of the olfactory nerve and olfactory bulb (OB) glomeruli begins embryonically in mice. However, the development of the olfactory system continues throughout life with the addition of new olfactory sensory neurons (OSNs) in the olfactory epithelium (OE). Much attention has been given to the perinatal innervation of the OB by OSN axons, but in the young adult the process of OSN maturation and axon targeting to the OB remains controversial. To address this gap in understanding, we used BrdU to label late-born OSNs in young adult mice at postnatal day 25 (P25-born OSNs) and timed their molecular maturation following basal cell division. We show that OSNs in young adults undergo a sequential molecular development with the expression of GAP 43 (growth-associated protein 43) > AC3 (adenylyl cyclase 3) > OMP (olfactory marker protein), consecutively, in a time frame of ∼8 d. To assess OSN axon development, we implemented an in vivo fate-mapping strategy to label P25-born OSNs with ZsGreen. Using sampling intervals of 24 h, we demonstrate the progressive extension of OSN axons in the OE, through the foramen of the cribriform plate, and onto the surface of the OB. OSN axons reached the OB and began to target and robustly innervate specific glomeruli ∼10 d following basal cell division, a time point at which OMP expression becomes evident. Our data demonstrate a sequential process of correlated axon extension and molecular maturation that is similar to that seen in the neonate, but on a slightly longer timescale and with regional differences in the OE.
Collapse
Affiliation(s)
- Teresa Liberia
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Eduardo Martin-Lopez
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Sarah J Meller
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06520
- The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Charles A Greer
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06520
- The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
16
|
Abstract
The formation of the olfactory nerve and olfactory bulb (OB) glomeruli begins embryonically in mice. However, the development of the olfactory system continues throughout life with the addition of new olfactory sensory neurons (OSNs) in the olfactory epithelium (OE). Much attention has been given to the perinatal innervation of the OB by OSN axons, but in the young adult the process of OSN maturation and axon targeting to the OB remains controversial. To address this gap in understanding, we used BrdU to label late-born OSNs in young adult mice at postnatal day 25 (P25-born OSNs) and timed their molecular maturation following basal cell division. We show that OSNs in young adults undergo a sequential molecular development with the expression of GAP 43 (growth-associated protein 43) > AC3 (adenylyl cyclase 3) > OMP (olfactory marker protein), consecutively, in a time frame of ∼8 d. To assess OSN axon development, we implemented an in vivo fate-mapping strategy to label P25-born OSNs with ZsGreen. Using sampling intervals of 24 h, we demonstrate the progressive extension of OSN axons in the OE, through the foramen of the cribriform plate, and onto the surface of the OB. OSN axons reached the OB and began to target and robustly innervate specific glomeruli ∼10 d following basal cell division, a time point at which OMP expression becomes evident. Our data demonstrate a sequential process of correlated axon extension and molecular maturation that is similar to that seen in the neonate, but on a slightly longer timescale and with regional differences in the OE.
Collapse
|
17
|
Degl’Innocenti A, Meloni G, Mazzolai B, Ciofani G. A purely bioinformatic pipeline for the prediction of mammalian odorant receptor gene enhancers. BMC Bioinformatics 2019; 20:474. [PMID: 31521109 PMCID: PMC6744719 DOI: 10.1186/s12859-019-3012-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 07/29/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND In most mammals, a vast array of genes coding for chemosensory receptors mediates olfaction. Odorant receptor (OR) genes generally constitute the largest multifamily (> 1100 intact members in the mouse). From the whole pool, each olfactory neuron expresses a single OR allele following poorly characterized mechanisms termed OR gene choice. OR genes are found in genomic aggregations known as clusters. Nearby enhancers, named elements, are crucial regulators of OR gene choice. Despite their importance, searching for new elements is burdensome. Other chemosensory receptor genes responsible for smell adhere to expression modalities resembling OR gene choice, and are arranged in genomic clusters - often with chromosomal linkage to OR genes. Still, no elements are known for them. RESULTS Here we present an inexpensive framework aimed at predicting elements. We redefine cluster identity by focusing on multiple receptor gene families at once, and exemplify thirty - not necessarily OR-exclusive - novel candidate enhancers. CONCLUSIONS The pipeline we introduce could guide future in vivo work aimed at discovering/validating new elements. In addition, our study provides an updated and comprehensive classification of all genomic loci responsible for the transduction of olfactory signals in mammals.
Collapse
Affiliation(s)
- Andrea Degl’Innocenti
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera (Pisa), Italy
- Max Planck Institute for Biophysics, Max-Planck-Gesellschaft, Max-von-Laue-Straße 3, D-60438 Frankfurt am Main, Germany
| | - Gabriella Meloni
- Center for Micro-BioRobotics, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera (Pisa), Italy
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Viale Rinaldo Piaggio 34, 56025 Pontedera (Pisa), Italy
| | - Barbara Mazzolai
- Center for Micro-BioRobotics, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera (Pisa), Italy
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera (Pisa), Italy
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
18
|
Santos PSC, Mezger M, Kolar M, Michler FU, Sommer S. The best smellers make the best choosers: mate choice is affected by female chemosensory receptor gene diversity in a mammal. Proc Biol Sci 2019; 285:20182426. [PMID: 30963892 DOI: 10.1098/rspb.2018.2426] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The products of the genes of the major histocompatibility complex (MHC) are known to be drivers of pathogen resistance and sexual selection enhancing offspring genetic diversity. The MHC further influences individual odour types and social communication. However, little is known about the receptors and their volatile ligands that are involved in this type of chemical communication. Here, we have investigated chemosensory receptor genes that ultimately enable females to assess male genes through odour cues. As a model, we used an invasive population of North American raccoons ( Procyon lotor) in Germany. We investigated the effect of two groups of chemosensory receptor genes-trace amine-associated receptors (TAARs) and olfactory receptors (ORs)-on MHC-dependent mate choice. Females with more alleles of the TAAR or OR loci were more likely to choose a male with a diverse MHC. We additionally found that MHC class I genes have a stronger effect on mate choice than the recently reported effect for MHC class II genes, probably because of their immunological relevance for viral resistance. Our study is among the first to show a genetic link between behaviour and chemosensory receptor genes. These results contribute to understanding the link between genetics, olfaction and associated life-history decisions.
Collapse
Affiliation(s)
- Pablo S C Santos
- 1 Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm , Ulm , Germany
| | - Maja Mezger
- 1 Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm , Ulm , Germany
| | - Miriam Kolar
- 1 Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm , Ulm , Germany
| | - Frank-Uwe Michler
- 2 Institute of Forest Botany and Forest Zoology, Technical University of Dresden , Tharandt , Germany
| | - Simone Sommer
- 1 Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm , Ulm , Germany
| |
Collapse
|
19
|
Abstract
Olfaction plays a critical role in several aspects of life. Olfactory disorders are very common in the general population, and can lead to malnutrition, weight loss, food poisoning, depression, and other disturbances. Odorants are first detected in the upper region of the nose by the main olfactory epithelium (OE). In this region, millions of olfactory sensory neurons (OSNs) interact with odor molecules through the odorant receptors (ORs), which belong to the superfamily of G protein-coupled receptors. The binding of odors to the ORs initiates an electrical signal that travels along the axons to the main olfactory bulb of the brain. The information is then transmitted to other regions of the brain, leading to odorant perception and emotional and behavioral responses. In the OE, OSNs die and are continuously replaced from stem cells localized in the epithelium's basal region. Damage to this epithelium can be caused by multiple factors, leading to anosmia (smell loss). In this chapter, we introduce the basic organization of the OE and focus on the molecular mechanisms involved in odorant perception. We also describe recent experiments that address the mechanisms of OSNs regeneration in response to neuronal injury.
Collapse
Affiliation(s)
- Isaías Glezer
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bettina Malnic
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
20
|
Ishii KK, Touhara K. Neural circuits regulating sexual behaviors via the olfactory system in mice. Neurosci Res 2018; 140:59-76. [PMID: 30389572 DOI: 10.1016/j.neures.2018.10.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/25/2018] [Accepted: 10/15/2018] [Indexed: 01/17/2023]
Abstract
Reproduction is essential for any animal species. Reproductive behaviors, or sexual behaviors, are largely shaped by external sensory cues exchanged during sexual interaction. In many animals, including rodents, olfactory cues play a critical role in regulating sexual behavior. What exactly these olfactory cues are and how they impact animal behavior have been a central question in the field. Over the past few decades, many studies have dedicated to identifying an active compound that elicits sexual behavior from crude olfactory components. The identified substance has served as a tool to dissect the sensory processing mechanisms in the olfactory systems. In addition, recent advances in genetic engineering, and optics and microscopic techniques have greatly expanded our knowledge of the neural mechanisms underlying the control of sexual behavior in mice. This review summarizes our current knowledge about how sexual behaviors are controlled by olfactory cues.
Collapse
Affiliation(s)
- Kentaro K Ishii
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan; ERATO Touhara Chemosensory Signal Project, JST, The University of Tokyo, Tokyo 113-8657, Japan
| | - Kazushige Touhara
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan; ERATO Touhara Chemosensory Signal Project, JST, The University of Tokyo, Tokyo 113-8657, Japan.
| |
Collapse
|
21
|
Meyer A, Gläser A, Bräuer AU, Wree A, Strotmann J, Rolfs A, Witt M. Olfactory Performance as an Indicator for Protective Treatment Effects in an Animal Model of Neurodegeneration. Front Integr Neurosci 2018; 12:35. [PMID: 30154701 PMCID: PMC6102364 DOI: 10.3389/fnint.2018.00035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Neurodegenerative diseases are often accompanied by olfactory deficits. Here we use a rare neurovisceral lipid storage disorder, Niemann–Pick disease C1 (NPC1), to illustrate disease-specific dynamics of olfactory dysfunction and its reaction upon therapy. Previous findings in a transgenic mouse model (NPC1-/-) showed severe morphological and electrophysiological alterations of the olfactory epithelium (OE) and the olfactory bulb (OB) that ameliorated under therapy with combined 2-hydroxypropyl-ß-cyclodextrin (HPßCD)/allopregnanolone/miglustat or HPßCD alone. Methods: A buried pellet test was conducted to assess olfactory performance. qPCR for olfactory key markers and several olfactory receptors was applied to determine if their expression was changed under treatment conditions. In order to investigate the cell dynamics of the OB, we determined proliferative and apoptotic activities using a bromodeoxyuridine (BrdU) protocol and caspase-3 (cas-3) activity. Further, we performed immunohistochemistry and western blotting for microglia (Iba1), astroglia (GFAP) and tyrosine hydroxylase (TH). Results: The buried pellet test revealed a significant olfactory deterioration in NPC1-/- mice, which reverted to normal levels after treatment. At the OE level, mRNA for olfactory markers showed no changes; the mRNA level of classical olfactory receptor (ORs) was unaltered, that of unique ORs was reduced. In the OB of untreated NPC1-/- mice, BrdU and cas-3 data showed increased proliferation and apoptotic activity, respectively. At the protein level, Iba1 and GFAP in the OB indicated increased microgliosis and astrogliosis, which was prevented by treatment. Conclusion: Due to the unique plasticity especially of peripheral olfactory components the results show a successful treatment in NPC1 condition with respect to normalization of olfaction. Unchanged mRNA levels for olfactory marker protein and distinct olfactory receptors indicate no effects in the OE in NPC1-/- mice. Olfactory deficits are thus likely due to central deficits at the level of the OB. Further studies are needed to examine if olfactory performance can also be changed at a later onset and interrupted treatment of the disease. Taken together, our results demonstrate that olfactory testing in patients with NPC1 may be successfully used as a biomarker during the monitoring of the treatment.
Collapse
Affiliation(s)
- Anja Meyer
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Anne Gläser
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Anja U Bräuer
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Jörg Strotmann
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Arndt Rolfs
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Rostock, Germany
| | - Martin Witt
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
22
|
Octura JER, Maeda KI, Wakabayashi Y. Structure and zonal expression of olfactory receptors in the olfactory epithelium of the goat, Capra hircus. J Vet Med Sci 2018; 80:913-920. [PMID: 29681556 PMCID: PMC6021871 DOI: 10.1292/jvms.17-0692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mammalian olfactory system employs sophisticated mechanisms to detect and recognize an extensive range of smells. In rodents, the olfactory epithelium (OE), situated within the nasal cavity, mainly comprises four defined endoturbinates and several ectoturbinates. Olfactory receptors (ORs) belong to a large family, comprising over 1,000 genes in rodents, which are expressed in olfactory sensory neurons in the OE that detect odor molecules. The rodent OE is divided into four topographically distinct zones, defined by individual OR distribution. However, although the structural complexity and the zonal organization of mammalian OE may contribute to successfully interpreting olfactory information, it remains poorly understood. In this study, we investigated the nasal cavity structure and zonal organization of the OE in goats. Morphological observations revealed that the goat nasal cavity possessed well-developed endoturbinates and ectoturbinates and had a structure similar to that of rodents and sheep, previously reported in other studies. In situ hybridization was used to analyze the expression pattern of ORs, NADPH:quinone oxidoreductase 1, and olfactory cell adhesion molecules as markers of zonal organization in the goat OE. Based on the expression patterns of these genes, we concluded that the goat OE was divided into four zones. The well-developed structure of the nasal cavity and distribution of each OR in the OE were similar to those found in rodents, suggesting that these features were highly conserved between mammals and may have fundamental roles in discriminating among numerous odor molecules in the environment.
Collapse
Affiliation(s)
- Josh Elisha R Octura
- Department of Veterinary Medical Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.,Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), 2 Ikenodai, Tsukuba, Ibaraki 305-0901, Japan
| | - Kei-Ichiro Maeda
- Department of Veterinary Medical Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yoshihiro Wakabayashi
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), 2 Ikenodai, Tsukuba, Ibaraki 305-0901, Japan
| |
Collapse
|
23
|
Coppola DM, Ritchie BE, Craven BA. Tests of the sorption and olfactory "fovea" hypotheses in the mouse. J Neurophysiol 2017; 118:2770-2788. [PMID: 28877965 DOI: 10.1152/jn.00455.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/07/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022] Open
Abstract
The spatial distribution of receptors within sensory epithelia (e.g., retina and skin) is often markedly nonuniform to gain efficiency in information capture and neural processing. By contrast, odors, unlike visual and tactile stimuli, have no obvious spatial dimension. What need then could there be for either nearest-neighbor relationships or nonuniform distributions of receptor cells in the olfactory epithelium (OE)? Adrian (Adrian ED. J Physiol 100: 459-473, 1942; Adrian ED. Br Med Bull 6: 330-332, 1950) provided the only widely debated answer to this question when he posited that the physical properties of odors, such as volatility and water solubility, determine a spatial pattern of stimulation across the OE that could aid odor discrimination. Unfortunately, despite its longevity, few critical tests of the "sorption hypothesis" exist. Here we test the predictions of this hypothesis by mapping mouse OE responses using the electroolfactogram (EOG) and comparing these response "maps" to computational fluid dynamics (CFD) simulations of airflow and odorant sorption patterns in the nasal cavity. CFD simulations were performed for airflow rates corresponding to quiet breathing and sniffing. Consistent with predictions of the sorption hypothesis, water-soluble odorants tended to evoke larger EOG responses in the central portion of the OE than the peripheral portion. However, sorption simulation patterns along individual nasal turbinates for particular odorants did not correlate with their EOG response gradients. Indeed, the most consistent finding was a rostral-greater to caudal-lesser response gradient for all the odorants tested that is unexplained by sorption patterns. The viability of the sorption and related olfactory "fovea" hypotheses are discussed in light of these findings.NEW & NOTEWORTHY Two classical ideas concerning olfaction's receptor-surface two-dimensional organization-the sorption and olfactory fovea hypotheses-were found wanting in this study that afforded unprecedented comparisons between electrophysiological recordings in the mouse olfactory epithelium and computational fluid dynamic simulations of nasal airflow. Alternatively, it is proposed that the olfactory receptor layouts in macrosmatic mammals may be an evolutionary contingent state devoid of the functional significance found in other sensory epithelia like the cochlea and retina.
Collapse
Affiliation(s)
| | | | - Brent A Craven
- Mechanical and Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
24
|
Degl'Innocenti A, D'Errico A. Regulatory Features for Odorant Receptor Genes in the Mouse Genome. Front Genet 2017; 8:19. [PMID: 28270833 PMCID: PMC5318403 DOI: 10.3389/fgene.2017.00019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/08/2017] [Indexed: 12/31/2022] Open
Abstract
The odorant receptor genes, seven transmembrane receptor genes constituting the vastest mammalian gene multifamily, are expressed monogenically and monoallelicaly in each sensory neuron in the olfactory epithelium. This characteristic, often referred to as the one neuron-one receptor rule, is driven by mostly uncharacterized molecular dynamics, generally named odorant receptor gene choice. Much attention has been paid by the scientific community to the identification of sequences regulating the expression of odorant receptor genes within their loci, where related genes are usually arranged in genomic clusters. A number of studies identified transcription factor binding sites on odorant receptor promoter sequences. Similar binding sites were also found on a number of enhancers that regulate in cis their transcription, but have been proposed to form interchromosomal networks. Odorant receptor gene choice seems to occur via the local removal of strongly repressive epigenetic markings, put in place during the maturation of the sensory neuron on each odorant receptor locus. Here we review the fast-changing state of art for the study of regulatory features for odorant receptor genes.
Collapse
Affiliation(s)
- Andrea Degl'Innocenti
- Max Planck Institute of BiophysicsFrankfurt am Main, Germany; Cell and Developmental Biology Unit, Department of Biology, University of PisaPisa, Italy; Center for Micro-BioRobotics, Italian Institute of Technology, Sant'Anna School of Advanced StudiesPisa, Italy
| | - Anna D'Errico
- Max Planck Institute of Biophysics Frankfurt am Main, Germany
| |
Collapse
|
25
|
Molinas A, Aoudé I, Soubeyre V, Tazir B, Cadiou H, Grosmaitre X. Anatomical and molecular consequences of Unilateral Naris Closure on two populations of olfactory sensory neurons expressing defined odorant receptors. Neurosci Lett 2016; 626:42-7. [PMID: 27189720 DOI: 10.1016/j.neulet.2016.05.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/29/2016] [Accepted: 05/13/2016] [Indexed: 12/17/2022]
Abstract
Mammalian olfactory sensory neurons (OSNs), the primary elements of the olfactory system, are located in the olfactory epithelium lining the nasal cavity. Exposed to the environment, their lifespan is short. Consequently, OSNs are regularly regenerated and several reports show that activity strongly modulates their development and regeneration: the peripheral olfactory system can adjust to the amount of stimulus through compensatory mechanisms. Unilateral naris occlusion (UNO) was frequently used to investigate this mechanism at the entire epithelium level. However, there is little data regarding the effects of UNO at the cellular level, especially on individual neuronal populations expressing a defined odorant receptor. Here, using UNO during the first three postnatal weeks, we analyzed the anatomical and molecular consequences of sensory deprivation in OSNs populations expressing the MOR23 and M71 receptors. The density of MOR23-expressing neurons is decreased in the closed side while UNO does not affect the density of M71-expressing neurons. Using Real Time qPCR on isolated neurons, we observed that UNO modulates the transcript levels for transduction pathway proteins (odorant receptors, CNGA2, PDE1c). The transcripts modulated by UNO will differ between populations depending on the receptor expressed. These results suggest that sensory deprivation will have different effects on different OSNs' populations. As a consequence, early experience will shape the functional properties of OSNs differently depending on the type of odorant receptor they express.
Collapse
Affiliation(s)
- Adrien Molinas
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Imad Aoudé
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Vanessa Soubeyre
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Bassim Tazir
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Hervé Cadiou
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Xavier Grosmaitre
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| |
Collapse
|
26
|
Kilinc S, Savarino A, Coleman JH, Schwob JE, Lane RP. Lysine-specific demethylase-1 (LSD1) is compartmentalized at nuclear chromocenters in early post-mitotic cells of the olfactory sensory neuronal lineage. Mol Cell Neurosci 2016; 74:58-70. [PMID: 26947098 DOI: 10.1016/j.mcn.2016.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 01/29/2016] [Accepted: 03/02/2016] [Indexed: 12/12/2022] Open
Abstract
Mammalian olfaction depends on the development of specialized olfactory sensory neurons (OSNs) that each express one odorant receptor (OR) protein from a large family of OR genes encoded in the genome. The lysine-specific demethylase-1 (LSD1) protein removes activating H3K4 or silencing H3K9 methylation marks at gene promoters and is required for proper OR regulation. We show that LSD1 protein exhibits variable organization within nuclei of developing OSNs, and tends to consolidate into a single dominant compartment at the edges of chromocenters within nuclei of early post-mitotic cells of the mouse olfactory epithelium (MOE). Using an immortalized cell line derived from developing olfactory placode, we show that consolidation of LSD1 appears to be cell-cycle regulated, with a peak occurrence in early G1. LSD1 co-compartmentalizes with CoREST, a protein known to collaborate with LSD1 to carry out a variety of chromatin-modifying functions. We show that LSD1 compartments co-localize with 1-3 OR loci at the exclusion of most OR genes, and commonly associate with Lhx2, a transcription factor involved in OR regulation. Together, our data suggests that LSD1 is sequestered into a distinct nuclear space that might restrict a histone-modifying function to a narrow developmental time window and/or range of OR gene targets.
Collapse
Affiliation(s)
- Seda Kilinc
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06457, USA.
| | - Alyssa Savarino
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06457, USA
| | - Julie H Coleman
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - James E Schwob
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Robert P Lane
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06457, USA.
| |
Collapse
|
27
|
Calfún C, Domínguez C, Pérez-Acle T, Whitlock KE. Changes in Olfactory Receptor Expression Are Correlated With Odor Exposure During Early Development in the zebrafish (Danio rerio). Chem Senses 2016; 41:301-12. [PMID: 26892307 DOI: 10.1093/chemse/bjw002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We have previously shown that exposure to phenyl ethyl alcohol (PEA) causes an increase in the expression of the transcription factor otx2 in the olfactory epithelium (OE) of juvenile zebrafish, and this change is correlated with the formation of an odor memory of PEA. Here, we show that the changes in otx2 expression are specific to βPEA: exposure to αPEA did not affect otx2 expression. We identified 34 olfactory receptors (ORs) representing 16 families on 4 different chromosomes as candidates for direct regulation of OR expression via Otx2. Subsequent in silico analysis uncovered Hnf3b binding sites closely associated with Otx2 binding sites in the regions flanking the ORs. Analysis by quantitative polymerase chain reaction and RNA-seq of OR expression in developing zebrafish exposed to different isoforms of PEA showed that a subset of ORs containing both Otx2/Hnf3b binding sites were downregulated only in βPEA-exposed juveniles and this change persisted through adult life. Localization of OR expression by in situ hybridization indicates the downregulation occurs at the level of RNA and not the number of cells expressing a given receptor. Finally, analysis of immediate early gene expression in the OE did not reveal changes in c-fos expression in response to either αPEA or βPEA.
Collapse
Affiliation(s)
- Cristian Calfún
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Pasaje Harrington 287, Valparaíso 2360102, Chile, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2360102, Chile
| | - Calixto Domínguez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Pasaje Harrington 287, Valparaíso 2360102, Chile, Center for Bioinformatics and Genome Biology, Fundación Ciencia & Vida, Av. Zañartu 1482, Santiago 7750000, Chile and
| | - Tomás Pérez-Acle
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Pasaje Harrington 287, Valparaíso 2360102, Chile, Computational Biology Lab, Fundación Ciencia & Vida, Av. Zañartu 1482, Santiago 7750000, Chile
| | - Kathleen E Whitlock
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Pasaje Harrington 287, Valparaíso 2360102, Chile, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2360102, Chile,
| |
Collapse
|
28
|
Degl'Innocenti A, Parrilla M, Harr B, Teschke M. The Mouse Solitary Odorant Receptor Gene Promoters as Models for the Study of Odorant Receptor Gene Choice. PLoS One 2016; 11:e0144698. [PMID: 26794459 PMCID: PMC4721658 DOI: 10.1371/journal.pone.0144698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In vertebrates, several anatomical regions located within the nasal cavity mediate olfaction. Among these, the main olfactory epithelium detects most conventional odorants. Olfactory sensory neurons, provided with cilia exposed to the air, detect volatile chemicals via an extremely large family of seven-transmembrane chemoreceptors named odorant receptors. Their genes are expressed in a monogenic and monoallelic fashion: a single allele of a single odorant receptor gene is transcribed in a given mature neuron, through a still uncharacterized molecular mechanism known as odorant receptor gene choice. AIM Odorant receptor genes are typically arranged in genomic clusters, but a few are isolated (we call them solitary) from the others within a region broader than 1 Mb upstream and downstream with respect to their transcript's coordinates. The study of clustered genes is problematic, because of redundancy and ambiguities in their regulatory elements: we propose to use the solitary genes as simplified models to understand odorant receptor gene choice. PROCEDURES Here we define number and identity of the solitary genes in the mouse genome (C57BL/6J), and assess the conservation of the solitary status in some mammalian orthologs. Furthermore, we locate their putative promoters, predict their homeodomain binding sites (commonly present in the promoters of odorant receptor genes) and compare candidate promoter sequences with those of wild-caught mice. We also provide expression data from histological sections. RESULTS In the mouse genome there are eight intact solitary genes: Olfr19 (M12), Olfr49, Olfr266, Olfr267, Olfr370, Olfr371, Olfr466, Olfr1402; five are conserved as solitary in rat. These genes are all expressed in the main olfactory epithelium of three-day-old mice. The C57BL/6J candidate promoter of Olfr370 has considerably varied compared to its wild-type counterpart. Within the putative promoter for Olfr266 a homeodomain binding site is predicted. As a whole, our findings favor Olfr266 as a model gene to investigate odorant receptor gene choice.
Collapse
Affiliation(s)
- Andrea Degl'Innocenti
- Max-Planck-Institut für Biophysik, Frankfurt am Main, Germany
- Unità di Biologia Cellulare e dello Sviluppo, Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Marta Parrilla
- Max-Planck-Institut für Biophysik, Frankfurt am Main, Germany
| | - Bettina Harr
- Abteilung Evolutionsgenetik, Max-Planck-Institut für Evolutionsbiologie, Plön, Germany
| | - Meike Teschke
- Abteilung Evolutionsgenetik, Max-Planck-Institut für Evolutionsbiologie, Plön, Germany
| |
Collapse
|
29
|
Abdus-Saboor I, Fleischmann A, Shykind B. Setting limits: maintaining order in a large gene family. Transcription 2015; 5:e28978. [PMID: 25764336 PMCID: PMC4215173 DOI: 10.4161/trns.28978] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Odorant receptor (OR) gene choice is a paradigmatic example of stochastic regulation in which olfactory neurons choose one OR from > 1,000 possibilities. Recent biochemical, mathematical, and in vivo findings have revealed key players, introduced new axes of control, and brought the core mechanisms of the process into sharper focus.
Collapse
Affiliation(s)
- Ishmail Abdus-Saboor
- a Weill Cornell Medical College in Qatar; Qatar Foundation-Education City; Doha, Qatar
| | | | | |
Collapse
|
30
|
Abstract
The senses provide a means by which data on the physical and chemical properties of the environment may be collected and meaningfully interpreted. Sensation begins at the periphery, where a multitude of different sensory cell types are activated by environmental stimuli as different as photons and odorant molecules. Stimulus sensitivity is due to expression of different cell surface sensory receptors, and therefore the receptive field of each sense is defined by the aggregate of expressed receptors in each sensory tissue. Here, we review current understanding on patterns of expression and modes of regulation of sensory receptors.
Collapse
|
31
|
Olfactory receptor genes expressed in distinct lineages are sequestered in different nuclear compartments. Proc Natl Acad Sci U S A 2015; 112:E2403-9. [PMID: 25897022 DOI: 10.1073/pnas.1506058112] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The olfactory system translates a vast array of volatile chemicals into diverse odor perceptions and innate behaviors. Odor detection in the mouse nose is mediated by 1,000 different odorant receptors (ORs) and 14 trace amine-associated receptors (TAARs). ORs are used in a combinatorial manner to encode the unique identities of myriad odorants. However, some TAARs appear to be linked to innate responses, raising questions about regulatory mechanisms that might segregate OR and TAAR expression in appropriate subsets of olfactory sensory neurons (OSNs). Here, we report that OSNs that express TAARs comprise at least two subsets that are biased to express TAARs rather than ORs. The two subsets are further biased in Taar gene choice and their distribution within the sensory epithelium, with each subset preferentially expressing a subgroup of Taar genes within a particular spatial domain in the epithelium. Our studies reveal one mechanism that may regulate the segregation of Olfr (OR) and Taar expression in different OSNs: the sequestration of Olfr and Taar genes in different nuclear compartments. Although most Olfr genes colocalize near large central heterochromatin aggregates in the OSN nucleus, Taar genes are located primarily at the nuclear periphery, coincident with a thin rim of heterochromatin. Taar-expressing OSNs show a shift of one Taar allele away from the nuclear periphery. Furthermore, examination of hemizygous mice with a single Taar allele suggests that the activation of a Taar gene is accompanied by an escape from the peripheral repressive heterochromatin environment to a more permissive interior chromatin environment.
Collapse
|
32
|
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
|
33
|
Abstract
The mammalian olfactory system detects a plethora of environmental chemicals that are perceived as odors or stimulate instinctive behaviors. Studies using odorant receptor (OR) genes have provided insight into the molecular and organizational strategies underlying olfaction in mice. One important unanswered question, however, is whether these strategies are conserved in primates. To explore this question, we examined the macaque, a higher primate phylogenetically close to humans. Here we report that the organization of sensory inputs in the macaque nose resembles that in mouse in some respects, but not others. As in mouse, neurons with different ORs are interspersed in the macaque nose, and there are spatial zones that differ in their complement of ORs and extend axons to different domains in the olfactory bulb of the brain. However, whereas the mouse has multiple discrete band-like zones, the macaque appears to have only two broad zones. It is unclear whether the organization of OR inputs in a rodent/primate common ancestor degenerated in primates or, alternatively became more sophisticated in rodents. The mouse nose has an additional small family of chemosensory receptors, called trace amine-associated receptors (TAARs), which may detect social cues. Here we find that TAARs are also expressed in the macaque nose, suggesting that TAARs may also play a role in human olfactory perception. We further find that one human TAAR responds to rotten fish, suggesting a possible role as a sentinel to discourage ingestion of food harboring pathogenic microorganisms.
Collapse
|
34
|
Markenscoff-Papadimitriou E, Allen WE, Colquitt BM, Goh T, Murphy KK, Monahan K, Mosley CP, Ahituv N, Lomvardas S. Enhancer interaction networks as a means for singular olfactory receptor expression. Cell 2014; 159:543-57. [PMID: 25417106 PMCID: PMC4243057 DOI: 10.1016/j.cell.2014.09.033] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 07/07/2014] [Accepted: 09/10/2014] [Indexed: 12/20/2022]
Abstract
The transcriptional activation of one out of ?2800 olfactory receptor (OR) alleles is a poorly understood process. Here, we identify a plethora of putative OR enhancers and study their in vivo activity in olfactory neurons. Distinguished by an unusual epigenetic signature, candidate OR enhancers are characterized by extensive interchromosomal interactions associated with OR transcription and share a similar pattern of transcription factor footprints. In particular, we establish the role of the transcription factor Bptf as a facilitator of both enhancer interactions and OR transcription. Our observations agree with the model whereby OR transcription occurs in the context of multiple interacting enhancers. Disruption of these interchromosomal interactions results in weak and multigenic OR expression, suggesting that the rare coincidence of numerous enhancers over a stochastically chosen OR may account for the singularity and robustness in OR transcription.
Collapse
Affiliation(s)
| | - William E Allen
- Neuroscience Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bradley M Colquitt
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tracie Goh
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Karl K Murphy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kevin Monahan
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Colleen P Mosley
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stavros Lomvardas
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
35
|
Lyons DB, Allen WE, Goh T, Tsai L, Barnea G, Lomvardas S. An epigenetic trap stabilizes singular olfactory receptor expression. Cell 2013; 154:325-36. [PMID: 23870122 PMCID: PMC3929589 DOI: 10.1016/j.cell.2013.06.039] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 05/06/2013] [Accepted: 06/20/2013] [Indexed: 11/27/2022]
Abstract
The molecular mechanisms regulating olfactory receptor (OR) expression in the mammalian nose are not yet understood. Here, we identify the transient expression of histone demethylase LSD1 and the OR-dependent expression of adenylyl cyclase 3 (Adcy3) as requirements for initiation and stabilization of OR expression. As a transcriptional coactivator, LSD1 is necessary for desilencing and initiating OR transcription, but as a transcriptional corepressor, it is incompatible with maintenance of OR expression, and its downregulation is imperative for stable OR choice. Adcy3, a sensor of OR expression and a transmitter of an OR-elicited feedback, mediates the downregulation of LSD1 and promotes the differentiation of olfactory sensory neurons (OSNs). This novel, three-node signaling cascade locks the epigenetic state of the chosen OR, stabilizes its singular expression, and prevents the transcriptional activation of additional OR alleles for the life of the neuron.
Collapse
Affiliation(s)
- David B Lyons
- Tetrad Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | | | | | |
Collapse
|
36
|
Dishevelled proteins are associated with olfactory sensory neuron presynaptic terminals. PLoS One 2013; 8:e56561. [PMID: 23437169 PMCID: PMC3577874 DOI: 10.1371/journal.pone.0056561] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 01/11/2013] [Indexed: 11/28/2022] Open
Abstract
Olfactory sensory neurons (OSNs) project their axons from the olfactory epithelium toward the olfactory bulb (OB) in a heterogeneous and unsorted arrangement. However, as the axons approach the glomerular layer of the OB, axons from OSNs expressing the same odorant receptor (OR) sort and converge to form molecularly homogeneous glomeruli. Axon guidance cues, cell adhesion molecules, and OR induced activity have been implicated in the final targeting of OSN axons to specific glomeruli. Less understood, and often controversial, are the mechanisms used by OSN axons to initially navigate from the OE toward the OB. We previously demonstrated a role for Wnt and Frizzled (Fz) molecules in OSN axon extension and organization within the olfactory nerve. Building on that we now turned our attention to the downstream signaling cascades from Wnt-Fz interactions. Dishevelled (Dvl) is a key molecule downstream of Fz receptors. Three isoforms of Dvl with specific as well as overlapping functions are found in mammals. Here, we show that Dvl-1 expression is restricted to OSNs in the dorsal recess of the nasal cavity, and labels a unique subpopulation of glomeruli. Dvl-2 and Dvl-3 have a widespread distribution in both the OE and OB. Both Dvl-1 and Dvl-2 are associated with intra-glomerular pre-synaptic OSN terminals, suggesting a role in synapse formation/stabilization. Moreover, because Dvl proteins were observed in all OSN axons, we hypothesize that they are important determinants of OSN cell differentiation and axon extension.
Collapse
|
37
|
Coppola DM, Waggener CT, Radwani SM, Brooks DA. An electroolfactogram study of odor response patterns from the mouse olfactory epithelium with reference to receptor zones and odor sorptiveness. J Neurophysiol 2013; 109:2179-91. [PMID: 23343905 DOI: 10.1152/jn.00769.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Olfactory sensory neuron (OSN) responses to odors, measured at the population level, tend to be spatially heterogeneous in the vertebrates that have been studied. These response patterns vary between odors but are similar across subjects for a given stimulus. However, few species have been studied making functional interpretation of these patterns problematic. One proximate explanation for the spatial heterogeneity of odor responses comes from evidence that olfactory receptor (OR) genes in rodents are expressed in OSN populations that are spatially restricted to a few zones in the olfactory epithelium (OE). A long-standing functional explanation for response anisotropy in the OE posits that it is the signature of a supplementary mechanism for quality coding, based on the sorptive properties of odor molecules. These theories are difficult to assess because most mapping studies have utilized few odors, provided little replication, or involved but a single species (rat). In fact, to our knowledge, a detailed olfactory response "map" has not been reported for mouse, the species used in most studies of gene localization. Here we report the results of a study of mouse OE response patterns using the electroolfactogram (EOG). We focused on the medial aspect of olfactory turbinates that are accessible in the midsagittal section. This limited approach still allowed us to test predictions derived from the zonal distribution of OSN types and the sorption hypothesis. In 3 separate experiments, 290 mice were used to record EOGs from a set of standard locations along each of 4 endoturbinates utilizing 11 different odors resulting in over 4,400 separate recordings. Our results confirmed a marked spatial heterogeneity in odor responses that varied with odor, as seen in other species. However, no discontinuities were found in the odor-specific response patterns across the OE as might have been predicted given the existence of classical receptor zones nor did we find clear support for the hypothesis that OE response patterns, presumably a reflection of OSN distribution, have been shaped through natural selection by the relative sorptive properties of odors. We propose that receptor zones may be an epiphenomenon of a contingent evolutionary process. In this formulation, constraints on developmental programs for distributing OSN classes within the OE may be minimally related to the odor ligands of specific class members. Further, we propose that odor sorptiveness, which appears to be correlated with the inherent response patterns in the OE of larger species, may be of minimal effect in mice owing to scaling issues.
Collapse
Affiliation(s)
- D M Coppola
- Dept. of Biology, Randolph Macon College, 304 Caroline St., Ashland, VA 23005, USA.
| | | | | | | |
Collapse
|
38
|
Santoro SW, Dulac C. The activity-dependent histone variant H2BE modulates the life span of olfactory neurons. eLife 2012; 1:e00070. [PMID: 23240083 PMCID: PMC3510456 DOI: 10.7554/elife.00070] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/18/2012] [Indexed: 12/17/2022] Open
Abstract
We have identified a replication-independent histone variant, Hist2h2be (referred to herein as H2be), which is expressed exclusively by olfactory chemosensory neurons. Levels of H2BE are heterogeneous among olfactory neurons, but stereotyped according to the identity of the co-expressed olfactory receptor (OR). Gain- and loss-of-function experiments demonstrate that changes in H2be expression affect olfactory function and OR representation in the adult olfactory epithelium. We show that H2BE expression is reduced by sensory activity and that it promotes neuronal cell death, such that inactive olfactory neurons display higher levels of the variant and shorter life spans. Post-translational modifications (PTMs) of H2BE differ from those of the canonical H2B, consistent with a role for H2BE in altering transcription. We propose a physiological function for H2be in modulating olfactory neuron population dynamics to adapt the OR repertoire to the environment. DOI:http://dx.doi.org/10.7554/eLife.00070.001 A hallmark of the nervous systems of all mammals is their capacity to undergo changes in function that are shaped by experience. This phenomenon underlies the ability of our brains to develop properly and to learn, and also enables various sensory systems—including the visual, auditory and olfactory systems—to perform optimally in diverse environments. In most mammals, a high-functioning olfactory system is essential for carrying out tasks that are crucial for survival, such as finding food, avoiding predators and mating. In general, sensory systems have to decipher only a limited collection of stimuli, but the olfactory system must be able to process information from thousands of distinct odors that are found in a given environment and which may vary dramatically from one environment to the next. Each odor-sensing neuron in the nose of a mammal contains just one kind of odorant receptor protein, although mammalian genomes typically encode 1000 or so different kinds of receptor proteins. This suggests that it might be possible to ‘tune’ the olfactory system to a particular environment by changing the relative numbers of the different types of neurons. Indeed, it is known that the relative abundance of each type of odor-sensing neuron changes with age and experience, and that these changes might be caused by variations in the lifespans of the neurons. Although our understanding of how these experience-dependent changes are orchestrated at the molecular level is far from complete, it is clear that adjustments in the levels of specific gene products is necessary. But how do experiences alter the levels of gene products to give rise to lasting changes in the brain? One hypothesis is that changes to a structure called chromatin are key to this process: chromatin is an assembly of DNA molecules, which are quite long, and organizing proteins, mostly proteins known as histones, that together form a compact structure that can fit inside the nucleus of a cell. Santoro and Dulac have now discovered a previously uncharacterized protein called H2BE that is found only in the odor-sensing neurons of mice. H2BE is a variant of a protein called H2B, which is a well-known histone. They found that in odor-sensing neurons, H2BE replaces H2B to an extent that depends on the amount of activity experienced by the neuron: H2BE is nearly undetectable in highly active neurons, but almost completely replaces H2B in neurons that are inactive. Moreover, genetic manipulation showed that the deletion of H2BE significantly extended the lifespan of neurons, whereas elevated levels of H2BE shortened their lifespan. These findings reveal an extraordinary process that involves inactive odor-sensing neurons being depleted relative to active ones over time. How does H2BE, which differs from H2B by just five amino acids, cause such dramatic changes in neuronal composition? One hint comes from evidence that these amino acids disrupt interactions between chromatin and ‘effector’ proteins, which modulate gene activity. Consistent with this, Santoro and Dulac have found that the replacement of H2B by H2BE strongly alters gene activity, although the precise mechanism by which these alterations regulate neuronal lifespans remains to be determined. Understanding this process in detail, and exploring if similar phenomena are involved in experience-dependent changes elsewhere in the nervous system, are fascinating areas of future research. DOI:http://dx.doi.org/10.7554/eLife.00070.002
Collapse
Affiliation(s)
- Stephen W Santoro
- Howard Hughes Medical Institute, Department of Molecular and Cellular Biology , Harvard University , Cambridge , United States
| | | |
Collapse
|
39
|
Blanco-Hernández E, Valle-Leija P, Zomosa-Signoret V, Drucker-Colín R, Vidaltamayo R. Odor memory stability after reinnervation of the olfactory bulb. PLoS One 2012; 7:e46338. [PMID: 23071557 PMCID: PMC3468571 DOI: 10.1371/journal.pone.0046338] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 08/31/2012] [Indexed: 11/19/2022] Open
Abstract
The olfactory system, particularly the olfactory epithelium, presents a unique opportunity to study the regenerative capabilities of the brain, because of its ability to recover after damage. In this study, we ablated olfactory sensory neurons with methimazole and followed the anatomical and functional recovery of circuits expressing genetic markers for I7 and M72 receptors (M72-IRES-tau-LacZ and I7-IRES-tau-GFP). Our results show that 45 days after methimazole-induced lesion, axonal projections to the bulb of M72 and I7 populations are largely reestablished. Furthermore, regenerated glomeruli are re-formed within the same areas as those of control, unexposed mice. This anatomical regeneration correlates with functional recovery of a previously learned odorant-discrimination task, dependent on the cognate ligands for M72 and I7. Following regeneration, mice also recover innate responsiveness to TMT and urine. Our findings show that regeneration of neuronal circuits in the olfactory system can be achieved with remarkable precision and underscore the importance of glomerular organization to evoke memory traces stored in the brain.
Collapse
Affiliation(s)
- Eduardo Blanco-Hernández
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Distrito Federal, México
| | - Pablo Valle-Leija
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Distrito Federal, México
| | - Viviana Zomosa-Signoret
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León, Nuevo León, México
| | - René Drucker-Colín
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Distrito Federal, México
| | - Román Vidaltamayo
- Departamento de Ciencias Básicas, Centro de Diagnóstico Molecular y Medicina Personalizada, Universidad de Monterrey, Nuevo León, México
- * E-mail:
| |
Collapse
|
40
|
Abstract
We constructed a soybean bacterial artificial chromosome (BAC) library suitable for map-based cloning and physical mapping in soybean. This library consists of approximately 40 000 clones (4-5 genome equivalents) stored individually in 384-well microtiter dishes. A random sampling of 224 clones yielded an average insert size of 150 kb, giving a 98% probability of recovering any specific sequence. We screened the library for seven single or very low copy genie or genomic sequences using the polymerase chain reaction (PCR) and found between one and seven BACs for each of the seven sequences. When testing the library with a portion of the soybean psbA chloroplast gene, we found less than 1% chloroplast DNA representation. We also screened the library for eight different classes of disease resistance gene analogs (RGAs) and identified BACs containing all RGAs except class 8. We arranged nine of the class 1 RGA BACs and six of the class 3 RGA BACs into individual contigs based on fingerprint patterns observed after Southern probing of restriction digests of the member BACs with a class-specific sequence. This resulted in the partial localization of the different multigene family sequences without precise definition of their exact positions. Using PCR-based end rescue techniques and RFLP mapping of BAC ends, we mapped individual BACs of each contig onto linkage group J of the soybean public map. The class 1 contig mapped to the region on linkage group J that contains several disease resistance genes. The class 1 contig extended approximately 400 kb. The arrangement of the BACs within this contig has been confirmed using PCR. One end of the class 1 contig core BAC mapped to two positions on linkage group J and cosegregated with two class 1 RGA loci, suggesting that this segment is within an area of regional duplication.
Collapse
|
41
|
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
|
42
|
Gutman DA, Magnuson M, Majeed W, Keifer OP, Davis M, Ressler KJ, Keilholz S. Mapping of the mouse olfactory system with manganese-enhanced magnetic resonance imaging and diffusion tensor imaging. Brain Struct Funct 2012; 218:527-37. [PMID: 22527121 DOI: 10.1007/s00429-012-0413-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 04/03/2012] [Indexed: 12/29/2022]
Abstract
As the power of studying mouse genetics and behavior advances, research tools to examine systems level connectivity in the mouse are critically needed. In this study, we compared statistical mapping of the olfactory system in adult mice using manganese-enhanced MRI (MEMRI) and diffusion tensor imaging (DTI) with probabilistic tractography. The primary goal was to determine whether these complementary techniques can determine mouse olfactory bulb (OB) connectivity consistent with known anatomical connections. For MEMRI, 3D T1-weighted images were acquired before and after bilateral nasal administration of MnCl(2) solution. Concomitantly, high-resolution diffusion-tensor images were obtained ex vivo from a second group of mice and processed with a probabilistic tractography algorithm originating in the OB. Incidence maps were created by co-registering and overlaying data from the two scan modalities. The resulting maps clearly show pathways between the OB and amygdala, piriform cortex, caudate putamen, and olfactory cortex in both the DTI and MEMRI techniques that are consistent with the known anatomical connections. These data demonstrate that MEMRI and DTI are complementary, high-resolution neuroimaging tools that can be applied to mouse genetic models of olfactory and limbic system connectivity.
Collapse
Affiliation(s)
- David A Gutman
- Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA 30329, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
Vedin V, Molander M, Bohm S, Berghard A. Regional differences in olfactory epithelial homeostasis in the adult mouse. J Comp Neurol 2009; 513:375-84. [PMID: 19177519 DOI: 10.1002/cne.21973] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The olfactory sensory neurons in the nasal cavity of the adult mouse are organized into a few regions that differ in their molecular properties, as several classes of genes show regional expression. Most renowned is the fact that expression of each of hundreds of different odorant receptor genes is limited to one such region, or zone, of the olfactory neuroepithelial sheet. Zone differences are in place at birth, as exemplified here by the expression of neuronal progenitor marker Foxg1. We herein describe that an adult pattern showing regional differences in neurogenesis develops during the first few weeks of postnatal life which, e.g., is reflected in the temporal and regional regulation of the neuronal progenitor marker Ascl1. The most dorsomedial zone shows significantly fewer cells in S-phase in the adult but not in newborn mice by two different measures. Moreover, we show that there are regional differences in the relative differentiation, cell survival, and thickness of the olfactory epithelium. These findings are compatible with the view that zones are inherently distinct and that such differences contribute to generate regional differences in cellular homeostasis that in turn may modulate the capacity of a region to adjust to extrinsic influence.
Collapse
Affiliation(s)
- Viktoria Vedin
- Department of Molecular Biology, Umeå University, Sweden
| | | | | | | |
Collapse
|
45
|
Furudono Y, Sone Y, Takizawa K, Hirono J, Sato T. Relationship between peripheral receptor code and perceived odor quality. Chem Senses 2008; 34:151-8. [PMID: 19073951 DOI: 10.1093/chemse/bjn071] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The discrimination of thousands of odorants is mediated by several hundred olfactory receptors (ORs). It is generally accepted that the main strategy in encoding odor quality is a combinatorial receptor code scheme, in which odorants are discriminated by different sets of ORs. In the present study, we classified 12 test odorants by their receptor codes and perceived odor qualities to examine whether odorants showing similar receptor codes are also similar in their odor qualities. Similarities of receptor codes between odorants were estimated by the overlapping responses of murine isolated olfactory sensory neurons. In contrast, we conducted a human sensory test to classify the test odorants according to their odor qualities. Despite the difference in species, the groupings of the test odorants were well conserved between receptor code and odor quality. These findings indicate that odorants that are discriminated by murine receptor codes are perceived as different odors by humans and further suggest that similarity of receptor codes correlates with that of odor quality, at least in our test odorants at the concentrations tested.
Collapse
Affiliation(s)
- Yuichi Furudono
- Tobacco Science Research Center, Japan Tobacco Inc., 6-2 Umegaoka, Yokohama, Aoba-ku, Kanagawa 227-8512, Japan.
| | | | | | | | | |
Collapse
|
46
|
Axon Guidance Events in the Wiring of the Mammalian Olfactory System. Mol Neurobiol 2008; 39:1-9. [DOI: 10.1007/s12035-008-8047-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 11/12/2008] [Indexed: 10/21/2022]
|
47
|
Haddad R, Carmel L, Sobel N, Harel D. Predicting the receptive range of olfactory receptors. PLoS Comput Biol 2008; 4:e18. [PMID: 18248088 PMCID: PMC2222922 DOI: 10.1371/journal.pcbi.0040018] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Accepted: 12/04/2007] [Indexed: 11/25/2022] Open
Abstract
Although the family of genes encoding for olfactory receptors was identified more than 15 years ago, the difficulty of functionally expressing these receptors in an heterologous system has, with only some exceptions, rendered the receptive range of given olfactory receptors largely unknown. Furthermore, even when successfully expressed, the task of probing such a receptor with thousands of odors/ligands remains daunting. Here we provide proof of concept for a solution to this problem. Using computational methods, we tune an electronic nose to the receptive range of an olfactory receptor. We then use this electronic nose to predict the receptors' response to other odorants. Our method can be used to identify the receptive range of olfactory receptors, and can also be applied to other questions involving receptor–ligand interactions in non-olfactory settings. A key goal in biology is to identify specific ligands for specific receptors. One example is where the ligand is a drug. In turn, in the olfactory system the ligand is the odorant that binds to olfactory receptors. There are many olfactory receptor types, and which odorants will activate which receptors remains largely unknown. One way to answer this is to systematically vary the molecular features of ligands and to measure the olfactory receptor response. However, the vast number of molecular features and their combinations renders such an effort potentially unsolvable. Here, rather than looking at the trees (each molecular feature), we looked at the forest (the smell they generate). We used a device called an electronic nose that generates a patterned response to odorants. We then obtained the response to a set of odorants that are known to activate a particular olfactory receptor, and we used this pattern to predict the response of that receptor to other odorants. We found that, on average in three out of four we could predict the response of olfactory receptors. This result provides a new method for probing the olfactory system, and also suggests a novel method for identifying potential drugs.
Collapse
Affiliation(s)
- Rafi Haddad
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel.
| | | | | | | |
Collapse
|
48
|
Abstract
The olfactory system meets niche- and species-specific demands by an accelerated evolution of its odorant receptor repertoires. In this review, we describe evolutionary processes that have shaped olfactory and vomeronasal receptor gene families in vertebrate genomes. We emphasize three important periods in the evolution of the olfactory system evident by comparative genomics: the adaptation to land in amphibian ancestors, the decline of olfaction in primates, and the delineation of putative pheromone receptors concurrent with rodent speciation. The rapid evolution of odorant receptor genes, the sheer size of the repertoire, as well as their wide distribution in the genome, presents a developmental challenge: how are these ever-changing odorant receptor repertoires coordinated within the olfactory system? A central organizing principle in olfaction is the specialization of sensory neurons resulting from each sensory neuron expressing only ~one odorant receptor allele. In this review, we also discuss this mutually exclusive expression of odorant receptor genes. We have considered several models to account for co-regulation of odorant receptor repertoires, as well as discussed a new hypothesis that invokes important epigenetic properties of the system.
Collapse
Affiliation(s)
- Marijo B Kambere
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Robert P Lane
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| |
Collapse
|
49
|
Cho JH, Lépine M, Andrews W, Parnavelas J, Cloutier JF. Requirement for Slit-1 and Robo-2 in zonal segregation of olfactory sensory neuron axons in the main olfactory bulb. J Neurosci 2007; 27:9094-104. [PMID: 17715346 PMCID: PMC6672192 DOI: 10.1523/jneurosci.2217-07.2007] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The formation of precise stereotypic connections in sensory systems is critical for the ability to detect and process signals from the environment. In the olfactory system, olfactory sensory neurons (OSNs) project axons to spatially defined glomeruli within the olfactory bulb (OB). A spatial relationship exists between the location of OSNs within the olfactory epithelium (OE) and their glomerular targets along the dorsoventral axis in the OB. The molecular mechanisms underlying the zonal segregation of OSN axons along the dorsoventral axis of the OB are poorly understood. Using robo-2(-/-) (roundabout) and slit-1(-/-) mice, we examined the role of the Slit family of axon guidance cues in the targeting of OSN axons during development. We show that a subset of OSN axons that normally project to the dorsal region of the OB mistarget and form glomeruli in the ventral region in robo-2(-/-) and slit-1(-/-) mice. In addition, we show that the Slit receptor, Robo-2, is expressed in OSNs in a high dorsomedial to low ventrolateral gradient across the OE and that Slit-1 and Slit-3 are expressed in the ventral region of the OB. These results indicate that the dorsal-to-ventral segregation of OSN axons are not solely defined by the location of OSNs within the OE but also relies on axon guidance cues.
Collapse
Affiliation(s)
- Jin Hyung Cho
- Montreal Neurological Institute, Centre for Neuronal Survival, Montréal, Québec, Canada H3A 2B4
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada H3A 2B4, and
| | - Manon Lépine
- Montreal Neurological Institute, Centre for Neuronal Survival, Montréal, Québec, Canada H3A 2B4
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada H3A 2B4, and
| | - William Andrews
- Medical Research Council Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | - John Parnavelas
- Medical Research Council Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | - Jean-François Cloutier
- Montreal Neurological Institute, Centre for Neuronal Survival, Montréal, Québec, Canada H3A 2B4
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada H3A 2B4, and
| |
Collapse
|
50
|
Sato Y, Miyasaka N, Yoshihara Y. Hierarchical regulation of odorant receptor gene choice and subsequent axonal projection of olfactory sensory neurons in zebrafish. J Neurosci 2007; 27:1606-15. [PMID: 17301169 PMCID: PMC6673750 DOI: 10.1523/jneurosci.4218-06.2007] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 12/18/2006] [Accepted: 01/08/2007] [Indexed: 11/21/2022] Open
Abstract
In both Drosophila and mice, olfactory sensory neurons (OSNs) expressing a given odorant receptor (OR) project axons to specific glomeruli in the antennal lobe or olfactory bulb (OB), developing a topographic odor map. To gain insights into the modes of OR expression and axonal projection in zebrafish, we generated a bacterial artificial chromosome transgenic line carrying an OR gene cluster in which two OR-coding sequences, OR111-7 and OR103-1, were replaced with yellow fluorescent protein (YFP) and cyan fluorescent protein (CFP), respectively. In the transgenic embryos, YFP and CFP signals appear in small populations of OSNs at an early stage of development when OR expression is first observed. Time-lapse imaging of living embryos revealed that both YFP- and CFP-expressing OSNs project axons to the medial portion of the OB. This pattern of axonal projection is maintained in the adult transgenic fish, in which fluorescently labeled OSN axons target a topographically fixed cluster of glomeruli in the medial OB. Because the OR-coding sequences were replaced with fluorescent reporter genes, we examined which OR genes are expressed in YFP/CFP-expressing OSNs and found that the OR choice is mostly restricted to OR members within the same subfamily of the cluster. Furthermore, we found that the one receptor-one neuron rule is not always applicable to zebrafish OSNs and that multiple receptors-one neuron is true for a subpopulation of OSNs in both wild-type and transgenic fish. These data demonstrate two distinct modes of OR expression and suggest a model of the hierarchical regulation of OR gene choice and subsequent axonal projection in the zebrafish olfactory system.
Collapse
Affiliation(s)
- Yuki Sato
- Laboratory for Neurobiology of Synapse, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan, and
| | - Nobuhiko Miyasaka
- Laboratory for Neurobiology of Synapse, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan, and
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Osaka 560-0082, Japan
| | - Yoshihiro Yoshihara
- Laboratory for Neurobiology of Synapse, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan, and
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Osaka 560-0082, Japan
| |
Collapse
|