1
|
Prakash N, Matos HY, Sebaoui S, Tsai L, Tran T, Aromolaran A, Atrachji I, Campbell N, Goodrich M, Hernandez-Pineda D, Jesus Herrero M, Hirata T, Lischinsky J, Martinez W, Torii S, Yamashita S, Hosseini H, Sokolowski K, Esumi S, Kawasawa YI, Hashimoto-Torii K, Jones KS, Corbin JG. Connectivity and molecular profiles of Foxp2- and Dbx1-lineage neurons in the accessory olfactory bulb and medial amygdala. J Comp Neurol 2024; 532:e25545. [PMID: 37849047 PMCID: PMC10922300 DOI: 10.1002/cne.25545] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023]
Abstract
In terrestrial vertebrates, the olfactory system is divided into main (MOS) and accessory (AOS) components that process both volatile and nonvolatile cues to generate appropriate behavioral responses. While much is known regarding the molecular diversity of neurons that comprise the MOS, less is known about the AOS. Here, focusing on the vomeronasal organ (VNO), the accessory olfactory bulb (AOB), and the medial amygdala (MeA), we reveal that populations of neurons in the AOS can be molecularly subdivided based on their ongoing or prior expression of the transcription factors Foxp2 or Dbx1, which delineate separate populations of GABAergic output neurons in the MeA. We show that a majority of AOB neurons that project directly to the MeA are of the Foxp2 lineage. Using single-neuron patch-clamp electrophysiology, we further reveal that in addition to sex-specific differences across lineage, the frequency of excitatory input to MeA Dbx1- and Foxp2-lineage neurons differs between sexes. Together, this work uncovers a novel molecular diversity of AOS neurons, and lineage and sex differences in patterns of connectivity.
Collapse
Affiliation(s)
- Nandkishore Prakash
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Heidi Y Matos
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Sonia Sebaoui
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Luke Tsai
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Tuyen Tran
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Adejimi Aromolaran
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Isabella Atrachji
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Nya Campbell
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Meredith Goodrich
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - David Hernandez-Pineda
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Maria Jesus Herrero
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Tsutomu Hirata
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Julieta Lischinsky
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Wendolin Martinez
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Shisui Torii
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Satoshi Yamashita
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Hassan Hosseini
- Department of Pharmacology, University of Michigan Medical
School, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan
Medical School, Ann Arbor, MI 48109, USA
| | - Katie Sokolowski
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Shigeyuki Esumi
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, PA, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Kevin S Jones
- Department of Pharmacology, University of Michigan Medical
School, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan
Medical School, Ann Arbor, MI 48109, USA
| | - Joshua G Corbin
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| |
Collapse
|
2
|
Zhang W, Huang H, Gui A, Mu D, Zhao T, Li H, Watanabe K, Xiao Z, Ye H, Xu Y. Contactin-6-deficient male mice exhibit the abnormal function of the accessory olfactory system and impaired reproductive behavior. Brain Behav 2023; 13:e2893. [PMID: 36860170 PMCID: PMC10097056 DOI: 10.1002/brb3.2893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 12/21/2022] [Accepted: 01/05/2023] [Indexed: 03/03/2023] Open
Abstract
INTRODUCTION Contactin-6 (CNTN6), also known as NB-3, is a neural recognition molecule and a member of the contactin subgroup of the immunoglobulin superfamily. Gene encoding CNTN6 is expressed in many regions of the neural system, including the accessory olfactory bulb (AOB) in mice. We aim to determine the effect of CNTN6 deficiency on the function of the accessory olfactory system (AOS). METHODS We examined the effect of CNTN6 deficiency on the reproductive behavior of male mice through behavioral experiments such as urine sniffing and mate preference tests. Staining and electron microscopy were used to observe the gross structure and the circuitry activity of the AOS. RESULTS Cntn6 is highly expressed in the vomeronasal organ (VNO) and the AOB, and sparsely expressed in the medial amygdala (MeA) and the medial preoptic area (MPOA), which receive direct and/or indirect projections from the AOB. Behavioral tests to examine reproductive function in mice, which is mostly controlled by the AOS, revealed that Cntn6-/- adult male mice showed less interest and reduced mating attempts toward estrous female mice in comparison with their Cntn6+/+ littermates. Although Cntn6-/- adult male mice displayed no obvious changes in the gross structure of the VNO or AOB, we observed the increased activation of granule cells in the AOB and the lower activation of neurons in the MeA and the MPOA as compared with Cntn6+/+ adult male mice. Moreover, there were an increased number of synapses between mitral cells and granule cells in the AOB of Cntn6-/- adult male mice as compared with wild-type controls. CONCLUSION These results indicate that CNTN6 deficiency affects the reproductive behavior of male mice, suggesting that CNTN6 participated in normal function of the AOS and its ablation was involved in synapse formation between mitral and granule cells in the AOB, rather than affecting the gross structure of the AOS.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Huiling Huang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Ailing Gui
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Di Mu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Tian Zhao
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Hongtao Li
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China
| | - Kazutada Watanabe
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata, Japan
| | - Zhicheng Xiao
- The Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Australia
| | - Haihong Ye
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Yiliang Xu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Kankanamge D, Tennakoon M, Karunarathne A, Gautam N. G protein gamma subunit, a hidden master regulator of GPCR signaling. J Biol Chem 2022; 298:102618. [PMID: 36272647 PMCID: PMC9678972 DOI: 10.1016/j.jbc.2022.102618] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/21/2022] Open
Abstract
Heterotrimeric G proteins (αβγ subunits) that are activated by G protein-coupled receptors (GPCRs) mediate the biological responses of eukaryotic cells to extracellular signals. The α subunits and the tightly bound βγ subunit complex of G proteins have been extensively studied and shown to control the activity of effector molecules. In contrast, the potential roles of the large family of γ subunits have been less studied. In this review, we focus on present knowledge about these proteins. Induced loss of individual γ subunit types in animal and plant models result in strikingly distinct phenotypes indicating that γ subtypes play important and specific roles. Consistent with these findings, downregulation or upregulation of particular γ subunit types result in various types of cancers. Clues about the mechanistic basis of γ subunit function have emerged from imaging the dynamic behavior of G protein subunits in living cells. This shows that in the basal state, G proteins are not constrained to the plasma membrane but shuttle between membranes and on receptor activation βγ complexes translocate reversibly to internal membranes. The translocation kinetics of βγ complexes varies widely and is determined by the membrane affinity of the associated γ subtype. On translocating, some βγ complexes act on effectors in internal membranes. The variation in translocation kinetics determines differential sensitivity and adaptation of cells to external signals. Membrane affinity of γ subunits is thus a parsimonious and elegant mechanism that controls information flow to internal cell membranes while modulating signaling responses.
Collapse
Affiliation(s)
- Dinesh Kankanamge
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Mithila Tennakoon
- Department of Chemistry, St Louis University, St Louis, Missouri, USA
| | | | - N Gautam
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA; Department of Genetics, Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
4
|
Lin JM, Mitchell TA, Rothstein M, Pehl A, Taroc EZM, Katreddi RR, Parra KE, Zuloaga DG, Simoes-Costa M, Forni PE. Sociosexual behavior requires both activating and repressive roles of Tfap2e/AP-2ε in vomeronasal sensory neurons. eLife 2022; 11:e77259. [PMID: 36111787 PMCID: PMC9525060 DOI: 10.7554/elife.77259] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Neuronal identity dictates the position in an epithelium, and the ability to detect, process, and transmit specific signals to specified targets. Transcription factors (TFs) determine cellular identity via direct modulation of genetic transcription and recruiting chromatin modifiers. However, our understanding of the mechanisms that define neuronal identity and their magnitude remain a critical barrier to elucidate the etiology of congenital and neurodegenerative disorders. The rodent vomeronasal organ provides a unique system to examine in detail the molecular mechanisms underlying the differentiation and maturation of chemosensory neurons. Here, we demonstrated that the identity of postmitotic/maturing vomeronasal sensory neurons (VSNs), and vomeronasal-dependent behaviors can be reprogrammed through the rescue of Tfap2e/AP-2ε expression in the Tfap2eNull mice, and partially reprogrammed by inducing ectopic Tfap2e expression in mature apical VSNs. We suggest that the TF Tfap2e can reprogram VSNs bypassing cellular plasticity restrictions, and that it directly controls the expression of batteries of vomeronasal genes.
Collapse
Affiliation(s)
- Jennifer M Lin
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Tyler A Mitchell
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Megan Rothstein
- Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
| | - Alison Pehl
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Ed Zandro M Taroc
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Raghu R Katreddi
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| | - Katherine E Parra
- Department of Psychology, University at Albany, State University of New YorkAlbanyUnited States
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, State University of New YorkAlbanyUnited States
| | - Marcos Simoes-Costa
- Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
| | - Paolo Emanuele Forni
- Department of Biological Sciences, University at Albany, State University of New YorkAlbanyUnited States
- The RNA Institute, University at AlbanyAlbanyUnited States
| |
Collapse
|
5
|
Merley AL, Hubbard JS, Rendahl AK, Boynton FDD, Impelluso LC. Behavioral and Physiologic Effects of Dirty Bedding Exposure in Female ICR Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2022; 61:42-51. [PMID: 34903313 PMCID: PMC8786380 DOI: 10.30802/aalas-jaalas-21-000060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/15/2021] [Accepted: 09/23/2021] [Indexed: 06/14/2023]
Abstract
Exposure of sentinel mice to dirty bedding is commonly used in health monitoring programs to screen colonies for clinical and subclinical disease. Despite the potential stressors present in dirty bedding, including but not limited to microorganisms, pheromones, and ammonia, it is unknown whether sentinel mice exposed to soiled bedding experience stress. In this study, select behavioral and physiologic changes associated with stress were assessed in female ICR mice exposed to dirty bedding. Behavioral parameters included evaluation in the home cage and selected behavioral tests; physiologic measurements included neutrophil:lymphocyte ratio and weight. Mice in the acute group were exposed for 24 h whereas mice in the chronic group were exposed for 4 wk. Mice in the chronic group exposed to dirty bedding weighed less at days 21 and 28 than did control mice. Chronic mice exposed to dirty bedding also exhibited decreased net weight gain over the entire study period as compared with control mice. No significant differences were detected in the other behavioral and physiologic parameters measured. These results indicate that dirty bedding exposure may affect sentinel mice, but further investigation is needed to determine the specific mechanism(s) behind the weight difference.
Collapse
Affiliation(s)
- Anne L Merley
- Research Animal Resources, University of Minnesota, Minneapolis, Minnesota
| | - Jennifer S Hubbard
- Research Animal Resources, University of Minnesota, Minneapolis, Minnesota
| | - Aaron K Rendahl
- College of Veterinary Medicine, University of Minnesota, Saint Paul, Minnesota
| | | | - Lynn Collura Impelluso
- Research Animal Resources, University of Minnesota, Minneapolis, Minnesota
- College of Veterinary Medicine, University of Minnesota, Saint Paul, Minnesota
| |
Collapse
|
6
|
Regulation of habenular G-protein gamma 8 on learning and memory via modulation of the central acetylcholine system. Mol Psychiatry 2021; 26:3737-3750. [PMID: 32989244 DOI: 10.1038/s41380-020-00893-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/24/2020] [Accepted: 09/15/2020] [Indexed: 01/19/2023]
Abstract
Guanine nucleotide binding protein (G protein) gamma 8 (Gng8) is a subunit of G proteins and expressed in the medial habenula (MHb) and interpeduncular nucleus (IPN). Recent studies have demonstrated that Gng8 is involved in brain development; however, the roles of Gng8 on cognitive function have not yet been addressed. In the present study, we investigated the expression of Gng8 in the brain and found that Gng8 was predominantly expressed in the MHb-IPN circuit of the mouse brain. We generated Gng8 knockout (KO) mice by CRISPR/Cas9 system in order to assess the role of Gng8 on cognitive function. Gng8 KO mice exhibited deficiency in learning and memory in passive avoidance and Morris water maze tests. In addition, Gng8 KO mice significantly reduced long-term potentiation (LTP) in the hippocampus compared to that of wild-type (WT) mice. Furthermore, we observed that levels of acetylcholine (ACh) and choline acetyltransferase (ChAT) in the MHb and IPN of Gng8 KO mice were significantly decreased, compared to WT mice. The administration of nAChR α4β2 agonist A85380 rescued memory impairment in the Gng8 KO mice, suggesting that Gng8 regulates cognitive function via modulation of cholinergic activity. Taken together, Gng8 is a potential therapeutic target for memory-related diseases and/or neurodevelopmental diseases.
Collapse
|
7
|
Manzini I, Schild D, Di Natale C. Principles of odor coding in vertebrates and artificial chemosensory systems. Physiol Rev 2021; 102:61-154. [PMID: 34254835 DOI: 10.1152/physrev.00036.2020] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The biological olfactory system is the sensory system responsible for the detection of the chemical composition of the environment. Several attempts to mimic biological olfactory systems have led to various artificial olfactory systems using different technical approaches. Here we provide a parallel description of biological olfactory systems and their technical counterparts. We start with a presentation of the input to the systems, the stimuli, and treat the interface between the external world and the environment where receptor neurons or artificial chemosensors reside. We then delineate the functions of receptor neurons and chemosensors as well as their overall I-O relationships. Up to this point, our account of the systems goes along similar lines. The next processing steps differ considerably: while in biology the processing step following the receptor neurons is the "integration" and "processing" of receptor neuron outputs in the olfactory bulb, this step has various realizations in electronic noses. For a long period of time, the signal processing stages beyond the olfactory bulb, i.e., the higher olfactory centers were little studied. Only recently there has been a marked growth of studies tackling the information processing in these centers. In electronic noses, a third stage of processing has virtually never been considered. In this review, we provide an up-to-date overview of the current knowledge of both fields and, for the first time, attempt to tie them together. We hope it will be a breeding ground for better information, communication, and data exchange between very related but so far little connected fields.
Collapse
Affiliation(s)
- Ivan Manzini
- Animal Physiology and Molecular Biomedicine, Justus-Liebig-University Gießen, Gießen, Germany
| | - Detlev Schild
- Institute of Neurophysiology and Cellular Biophysics, University Medical Center, University of Göttingen, Göttingen, Germany
| | - Corrado Di Natale
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
8
|
Katreddi RR, Forni PE. Mechanisms underlying pre- and postnatal development of the vomeronasal organ. Cell Mol Life Sci 2021; 78:5069-5082. [PMID: 33871676 PMCID: PMC8254721 DOI: 10.1007/s00018-021-03829-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/17/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
The vomeronasal organ (VNO) is sensory organ located in the ventral region of the nasal cavity in rodents. The VNO develops from the olfactory placode during the secondary invagination of olfactory pit. The embryonic vomeronasal structure appears as a neurogenic area where migratory neuronal populations like endocrine gonadotropin-releasing hormone-1 (GnRH-1) neurons form. Even though embryonic vomeronasal structures are conserved across most vertebrate species, many species including humans do not have a functional VNO after birth. The vomeronasal epithelium (VNE) of rodents is composed of two major types of vomeronasal sensory neurons (VSNs): (1) VSNs distributed in the apical VNE regions that express vomeronasal type-1 receptors (V1Rs) and the G protein subunit Gαi2, and (2) VSNs in the basal territories of the VNE that express vomeronasal type-2 receptors (V2Rs) and the G subunit Gαo. Recent studies identified a third subclass of Gαi2 and Gαo VSNs that express the formyl peptide receptor family. VSNs expressing V1Rs or V2Rs send their axons to distinct regions of the accessory olfactory bulb (AOB). Together, VNO and AOB form the accessory olfactory system (AOS), an olfactory subsystem that coordinates the social and sexual behaviors of many vertebrate species. In this review, we summarize our current understanding of cellular and molecular mechanisms that underlie VNO development. We also discuss open questions for study, which we suggest will further enhance our understanding of VNO morphogenesis at embryonic and postnatal stages.
Collapse
Affiliation(s)
- Raghu Ram Katreddi
- Department of Biological Sciences, Center for Neuroscience Research, The RNA Institute, University At Albany, State University of New York, Albany, NY, USA
| | - Paolo E Forni
- Department of Biological Sciences, Center for Neuroscience Research, The RNA Institute, University At Albany, State University of New York, Albany, NY, USA.
| |
Collapse
|
9
|
Tirindelli R. Coding of pheromones by vomeronasal receptors. Cell Tissue Res 2021; 383:367-386. [PMID: 33433690 DOI: 10.1007/s00441-020-03376-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/02/2020] [Indexed: 01/11/2023]
Abstract
Communication between individuals is critical for species survival, reproduction, and expansion. Most terrestrial species, with the exception of humans who predominantly use vision and phonation to create their social network, rely on the detection and decoding of olfactory signals, which are widely known as pheromones. These chemosensory cues originate from bodily fluids, causing attractive or avoidance behaviors in subjects of the same species. Intraspecific pheromone signaling is then crucial to identify sex, social ranking, individuality, and health status, thus establishing hierarchies and finalizing the most efficient reproductive strategies. Indeed, all these features require fine tuning of the olfactory systems to detect molecules containing this information. To cope with this complexity of signals, tetrapods have developed dedicated olfactory subsystems that refer to distinct peripheral sensory detectors, called the main olfactory and the vomeronasal organ, and two minor structures, namely the septal organ of Masera and the Grueneberg ganglion. Among these, the vomeronasal organ plays the most remarkable role in pheromone coding by mediating several behavioral outcomes that are critical for species conservation and amplification. In rodents, this organ is organized into two segregated neuronal subsets that express different receptor families. To some extent, this dichotomic organization is preserved in higher projection areas of the central nervous system, suggesting, at first glance, distinct functions for these two neuronal pathways. Here, I will specifically focus on this issue and discuss the role of vomeronasal receptors in mediating important innate behavioral effects through the recognition of pheromones and other biological chemosignals.
Collapse
Affiliation(s)
- Roberto Tirindelli
- Department of Medicine and Surgery, University of Parma, Via Volturno, 39, 43125, Parma, Italy.
| |
Collapse
|
10
|
Naik AS, Lin JM, Taroc EZM, Katreddi RR, Frias JA, Lemus AA, Sammons MA, Forni PE. Smad4-dependent morphogenic signals control the maturation and axonal targeting of basal vomeronasal sensory neurons to the accessory olfactory bulb. Development 2020; 147:147/8/dev184036. [PMID: 32341026 PMCID: PMC7197725 DOI: 10.1242/dev.184036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/10/2020] [Indexed: 12/31/2022]
Abstract
The vomeronasal organ (VNO) contains two main types of vomeronasal sensory neurons (VSNs) that express distinct vomeronasal receptor (VR) genes and localize to specific regions of the neuroepithelium. Morphogenic signals are crucial in defining neuronal identity and network formation; however, if and what signals control maturation and homeostasis of VSNs is largely unexplored. Here, we found transforming growth factor β (TGFβ) and bone morphogenetic protein (BMP) signal transduction in postnatal mice, with BMP signaling being restricted to basal VSNs and at the marginal zones of the VNO: the site of neurogenesis. Using different Smad4 conditional knockout mouse models, we disrupted canonical TGFβ/BMP signaling in either maturing basal VSNs (bVSNs) or all mature VSNs. Smad4 loss of function in immature bVSNs compromises dendritic knob formation, pheromone induced activation, correct glomeruli formation in the accessory olfactory bulb (AOB) and survival. However, Smad4 loss of function in all mature VSNs only compromises correct glomeruli formation in the posterior AOB. Our results indicate that Smad4-mediated signaling drives the functional maturation and connectivity of basal VSNs. Summary: Genetic disruption of TGFβ/BMP signaling in maturing basal vomeronasal sensory neurons (VSNs) or in all mature VSNs indicates that Smad4 signaling drives maturation and connectivity of basal VSNs.
Collapse
Affiliation(s)
- Ankana S Naik
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jennifer M Lin
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Ed Zandro M Taroc
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Raghu R Katreddi
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jesus A Frias
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Alex A Lemus
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Paolo E Forni
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| |
Collapse
|
11
|
Mohrhardt J, Nagel M, Fleck D, Ben-Shaul Y, Spehr M. Signal Detection and Coding in the Accessory Olfactory System. Chem Senses 2019; 43:667-695. [PMID: 30256909 PMCID: PMC6211456 DOI: 10.1093/chemse/bjy061] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In many mammalian species, the accessory olfactory system plays a central role in guiding behavioral and physiological responses to social and reproductive interactions. Because of its relatively compact structure and its direct access to amygdalar and hypothalamic nuclei, the accessory olfactory pathway provides an ideal system to study sensory control of complex mammalian behavior. During the last several years, many studies employing molecular, behavioral, and physiological approaches have significantly expanded and enhanced our understanding of this system. The purpose of the current review is to integrate older and newer studies to present an updated and comprehensive picture of vomeronasal signaling and coding with an emphasis on early accessory olfactory system processing stages. These include vomeronasal sensory neurons in the vomeronasal organ, and the circuitry of the accessory olfactory bulb. Because the overwhelming majority of studies on accessory olfactory system function employ rodents, this review is largely focused on this phylogenetic order, and on mice in particular. Taken together, the emerging view from both older literature and more recent studies is that the molecular, cellular, and circuit properties of chemosensory signaling along the accessory olfactory pathway are in many ways unique. Yet, it has also become evident that, like the main olfactory system, the accessory olfactory system also has the capacity for adaptive learning, experience, and state-dependent plasticity. In addition to describing what is currently known about accessory olfactory system function and physiology, we highlight what we believe are important gaps in our knowledge, which thus define exciting directions for future investigation.
Collapse
Affiliation(s)
- Julia Mohrhardt
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| | - Maximilian Nagel
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| | - David Fleck
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| | - Yoram Ben-Shaul
- Department of Medical Neurobiology, School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
12
|
Abstract
Most olfactory receptors in vertebrates are G protein-coupled receptors, whose activation by odorants initiates intracellular signaling cascades through heterotrimeric G proteins consisting of α, β, and γ subunits. Abolishment of the α subunits such as Gαolf in the main olfactory epithelium and Gαi2 and Gαo in the vomeronasal organ resulted in anosmia and/or impaired behavioral responses. In this study, we report that a G protein γ subunit, Gγ13, is expressed in a spatiotemporal manner similar to those of Gαolf and Gαi2 in the olfactory system and vomeronasal organ, respectively. In addition, Gγ13 was found in the glomeruli of the main olfactory bulb but was largely absent in the glomeruli of the accessory olfactory bulb. Using the Cre-loxP system, the Gγ13's gene Gng13 was nullified in the mature olfactory sensory neurons and apical vomeronasal sensory neurons where the Cre recombinase was expressed under the promoter of the Omp gene for the olfactory marker protein. Immunohistochemistry indicated much reduced expression of Gγ13 in the apical vomeronasal epithelium of the mutant mice. Behavioral experiments showed that the frequency and duration of aggressive encounters in the male mutant mice were significantly lower than in WT male mice. Taken together, these data suggest that the Gγ13 subunit is a critical signaling component in both the main olfactory epithelium and apical vomeronasal epithelium, and it plays an essential role in odor-triggered social behaviors including male-male aggression.
Collapse
|
13
|
Jager A, Maas DA, Fricke K, de Vries RB, Poelmans G, Glennon JC. Aggressive behavior in transgenic animal models: A systematic review. Neurosci Biobehav Rev 2018; 91:198-217. [DOI: 10.1016/j.neubiorev.2017.09.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/10/2017] [Accepted: 09/19/2017] [Indexed: 11/25/2022]
|
14
|
Lin JM, Taroc EZM, Frias JA, Prasad A, Catizone AN, Sammons MA, Forni PE. The transcription factor Tfap2e/AP-2ε plays a pivotal role in maintaining the identity of basal vomeronasal sensory neurons. Dev Biol 2018; 441:67-82. [PMID: 29928868 DOI: 10.1016/j.ydbio.2018.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/22/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022]
Abstract
The identity of individual neuronal cell types is defined and maintained by the expression of specific combinations of transcriptional regulators that control cell type-specific genetic programs. The epithelium of the vomeronasal organ of mice contains two major types of vomeronasal sensory neurons (VSNs): 1) the apical VSNs which express vomeronasal 1 receptors (V1r) and the G-protein subunit Gαi2 and; 2) the basal VSNs which express vomeronasal 2 receptors (V2r) and the G-protein subunit Gαo. Both cell types originate from a common pool of progenitors and eventually acquire apical or basal identity through largely unknown mechanisms. The transcription factor AP-2ε, encoded by the Tfap2e gene, plays a role in controlling the development of GABAergic interneurons in the main and accessory olfactory bulb (AOB), moreover AP-2ε has been previously described to be expressed in the basal VSNs. Here we show that AP-2ε is expressed in post-mitotic VSNs after they commit to the basal differentiation program. Loss of AP-2ε function resulted in reduced number of basal VSNs and in an increased number of neurons expressing markers of the apical lineage. Our work suggests that AP-2ε, which is expressed in late phases of differentiation, is not needed to initiate the apical-basal differentiation dichotomy but for maintaining the basal VSNs' identity. In AP-2ε mutants we observed a large number of cells that entered the basal program can express apical genes, our data suggest that differentiated VSNs of mice retain a notable level of plasticity.
Collapse
Affiliation(s)
- Jennifer M Lin
- Department of Biological Sciences, University at Albany, Albany, NY 12222, USA
| | - Ed Zandro M Taroc
- Department of Biological Sciences, University at Albany, Albany, NY 12222, USA
| | - Jesus A Frias
- Department of Biological Sciences, University at Albany, Albany, NY 12222, USA
| | - Aparna Prasad
- Department of Biological Sciences, University at Albany, Albany, NY 12222, USA
| | - Allison N Catizone
- Department of Biological Sciences, University at Albany, Albany, NY 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences, University at Albany, Albany, NY 12222, USA
| | - Paolo E Forni
- Department of Biological Sciences, University at Albany, Albany, NY 12222, USA.
| |
Collapse
|
15
|
Burger LL, Vanacker C, Phumsatitpong C, Wagenmaker ER, Wang L, Olson DP, Moenter SM. Identification of Genes Enriched in GnRH Neurons by Translating Ribosome Affinity Purification and RNAseq in Mice. Endocrinology 2018; 159. [PMID: 29522155 PMCID: PMC6287592 DOI: 10.1210/en.2018-00001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are a nexus of fertility regulation. We used translating ribosome affinity purification coupled with RNA sequencing to examine messenger RNAs of GnRH neurons in adult intact and gonadectomized (GDX) male and female mice. GnRH neuron ribosomes were tagged with green fluorescent protein (GFP) and GFP-labeled polysomes isolated by immunoprecipitation, producing one RNA fraction enhanced for GnRH neuron transcripts and one RNA fraction depleted. Complementary DNA libraries were created from each fraction and 50-base, paired-end sequencing done and differential expression (enhanced fraction/depleted fraction) determined with a threshold of >1.5- or <0.66-fold (false discovery rate P ≤ 0.05). A core of ∼840 genes was differentially expressed in GnRH neurons in all treatments, including enrichment for Gnrh1 (∼40-fold), and genes critical for GnRH neuron and/or gonadotrope development. In contrast, non-neuronal transcripts were not enriched or were de-enriched. Several epithelial markers were also enriched, consistent with the olfactory epithelial origins of GnRH neurons. Interestingly, many synaptic transmission pathways were de-enriched, in accordance with relatively low innervation of GnRH neurons. The most striking difference between intact and GDX mice of both sexes was a marked downregulation of genes associated with oxidative phosphorylation and upregulation of glucose transporters in GnRH neurons from GDX mice. This may suggest that GnRH neurons switch to an alternate fuel to increase adenosine triphosphate production in the absence of negative feedback when GnRH release is elevated. Knowledge of the GnRH neuron translatome and its regulation can guide functional studies and can be extended to disease states, such as polycystic ovary syndrome.
Collapse
Affiliation(s)
- Laura L Burger
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
| | - Charlotte Vanacker
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
| | | | - Elizabeth R Wagenmaker
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
| | - Luhong Wang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
| | - David P Olson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, Michigan
- Department of Internal Medicine, University of Michigan, Ann Arbor,
Michigan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor,
Michigan
- Correspondence: Laura L. Burger, PhD, University of Michigan, 7725 Med Sci II, 1137 E. Catherine
Street, Ann Arbor, Michigan 48109-5622. E-mail:
| |
Collapse
|
16
|
Jing YH, Qi CC, Yuan L, Liu XW, Gao LP, Yin J. Adult neural stem cell dysfunction in the subventricular zone of the lateral ventricle leads to diabetic olfactory defects. Neural Regen Res 2017; 12:1111-1118. [PMID: 28852393 PMCID: PMC5558490 DOI: 10.4103/1673-5374.211190] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Sensitive smell discrimination is based on structural plasticity of the olfactory bulb, which depends on migration and integration of newborn neurons from the subventricular zone. In this study, we examined the relationship between neural stem cell status in the subventricular zone and olfactory function in rats with diabetes mellitus. Streptozotocin was injected through the femoral vein to induce type 1 diabetes mellitus in Sprague-Dawley rats. Two months after injection, olfactory sensitivity was decreased in diabetic rats. Meanwhile, the number of BrdU-positive and BrdU+/DCX+ double-labeled cells was lower in the subventricular zone of diabetic rats compared with age-matched normal rats. Western blot results revealed downregulated expression of insulin receptor β, phosphorylated glycogen synthase kinase 3β, and β-catenin in the subventricular zone of diabetic rats. Altogether, these results indicate that diabetes mellitus causes insulin deficiency, which negatively regulates glycogen synthase kinase 3β and enhances β-catenin degradation, with these changes inhibiting neural stem cell proliferation. Further, these signaling pathways affect proliferation and differentiation of neural stem cells in the subventricular zone. Dysfunction of subventricular zone neural stem cells causes a decline in olfactory bulb structural plasticity and impairs olfactory sensitivity in diabetic rats.
Collapse
Affiliation(s)
- Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu Province, China
| | - Chu-Chu Qi
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Li Yuan
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Xiang-Wen Liu
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Jie Yin
- Institute of Anatomy and Histology & Embryology and Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
17
|
Beny Y, Kimchi T. Conditioned odor aversion induces social anxiety towards females in wild-type and TrpC2 knockout male mice. GENES BRAIN AND BEHAVIOR 2016; 15:722-732. [PMID: 27535696 DOI: 10.1111/gbb.12320] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/14/2016] [Accepted: 08/15/2016] [Indexed: 01/08/2023]
Abstract
Female-emitted pheromonal inputs possess an intrinsic rewarding value for conspecific males, promoting approach and investigation of the potential mating partner. In mice these inputs are detected mainly by the vomeronasal organ (VNO) and the main olfactory epithelium (MOE). We investigated the role of VNO-mediated inputs in experience-dependent plasticity of reproductive responses. We applied a sex-specific conditioned odor aversion (COA) paradigm on adult, wild-type (WT) male mice and on male mice impaired in VNO-mediated signal transduction (TrpC2-/- ). We found that WT males, which underwent COA to female-soiled bedding, lost their innate preference to female odors and presented lower motivation to approach a sexually receptive female. COA also abolished the testosterone surge normally seen following exposure to female odors. Moreover, the conditioned males displayed impairments in copulatory behaviors, which lasted for several weeks. Surprisingly, these males also exhibited phobic behaviors towards receptive females, including freezing and fleeing responses. In contrast, WT males which underwent COA specifically to male pheromones showed no change in olfactory preference and only a marginally significant elevation in intermale aggression. Finally, we show that TrpC2-/- males were able to acquire aversion to female-soiled bedding and presented similar behavioral alterations following COA in their responses to female cues. Our results demonstrate that the intrinsic rewarding value of female pheromones can be overridden through associative olfactory learning, which occurs independently of VNO inputs, probably through MOE signaling.
Collapse
Affiliation(s)
- Y Beny
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - T Kimchi
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
18
|
Freudenberg F, Carreño Gutierrez H, Post AM, Reif A, Norton WHJ. Aggression in non-human vertebrates: Genetic mechanisms and molecular pathways. Am J Med Genet B Neuropsychiatr Genet 2016; 171:603-40. [PMID: 26284957 DOI: 10.1002/ajmg.b.32358] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/28/2015] [Indexed: 11/07/2022]
Abstract
Aggression is an adaptive behavioral trait that is important for the establishment of social hierarchies and competition for mating partners, food, and territories. While a certain level of aggression can be beneficial for the survival of an individual or species, abnormal aggression levels can be detrimental. Abnormal aggression is commonly found in human patients with psychiatric disorders. The predisposition to aggression is influenced by a combination of environmental and genetic factors and a large number of genes have been associated with aggression in both human and animal studies. In this review, we compare and contrast aggression studies in zebrafish and mouse. We present gene ontology and pathway analyses of genes linked to aggression and discuss the molecular pathways that underpin agonistic behavior in these species. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | | | - Antonia M Post
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | - William H J Norton
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| |
Collapse
|
19
|
Veroude K, Zhang-James Y, Fernàndez-Castillo N, Bakker MJ, Cormand B, Faraone SV. Genetics of aggressive behavior: An overview. Am J Med Genet B Neuropsychiatr Genet 2016; 171B:3-43. [PMID: 26345359 DOI: 10.1002/ajmg.b.32364] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 08/05/2015] [Indexed: 12/24/2022]
Abstract
The Research Domain Criteria (RDoC) address three types of aggression: frustrative non-reward, defensive aggression and offensive/proactive aggression. This review sought to present the evidence for genetic underpinnings of aggression and to determine to what degree prior studies have examined phenotypes that fit into the RDoC framework. Although the constructs of defensive and offensive aggression have been widely used in the animal genetics literature, the human literature is mostly agnostic with regard to all the RDoC constructs. We know from twin studies that about half the variance in behavior may be explained by genetic risk factors. This is true for both dimensional, trait-like, measures of aggression and categorical definitions of psychopathology. The non-shared environment seems to have a moderate influence with the effects of shared environment being unclear. Human molecular genetic studies of aggression are in an early stage. The most promising candidates are in the dopaminergic and serotonergic systems along with hormonal regulators. Genome-wide association studies have not yet achieved genome-wide significance, but current samples are too small to detect variants having the small effects one would expect for a complex disorder. The strongest molecular evidence for a genetic basis for aggression comes from animal models comparing aggressive and non-aggressive strains or documenting the effects of gene knockouts. Although we have learned much from these prior studies, future studies should improve the measurement of aggression by using a systematic method of measurement such as that proposed by the RDoC initiative.
Collapse
Affiliation(s)
- Kim Veroude
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Yanli Zhang-James
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York.,Departments of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Catalonia, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Catalonia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain
| | - Mireille J Bakker
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Bru Cormand
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Catalonia, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Catalonia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain
| | - Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York.,Departments of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York.,K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| |
Collapse
|
20
|
Brignall AC, Cloutier JF. Neural map formation and sensory coding in the vomeronasal system. Cell Mol Life Sci 2015; 72:4697-709. [PMID: 26329476 PMCID: PMC11113928 DOI: 10.1007/s00018-015-2029-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/31/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022]
Abstract
Sensory systems enable us to encode a clear representation of our environment in the nervous system by spatially organizing sensory stimuli being received. The organization of neural circuitry to form a map of sensory activation is critical for the interpretation of these sensory stimuli. In rodents, social communication relies strongly on the detection of chemosignals by the vomeronasal system, which regulates a wide array of behaviours, including mate recognition, reproduction, and aggression. The binding of these chemosignals to receptors on vomeronasal sensory neurons leads to activation of second-order neurons within glomeruli of the accessory olfactory bulb. Here, vomeronasal receptor activation by a stimulus is organized into maps of glomerular activation that represent phenotypic qualities of the stimuli detected. Genetic, electrophysiological and imaging studies have shed light on the principles underlying cell connectivity and sensory map formation in the vomeronasal system, and have revealed important differences in sensory coding between the vomeronasal and main olfactory system. In this review, we summarize the key factors and mechanisms that dictate circuit formation and sensory coding logic in the vomeronasal system, emphasizing differences with the main olfactory system. Furthermore, we discuss how detection of chemosignals by the vomeronasal system regulates social behaviour in mice, specifically aggression.
Collapse
Affiliation(s)
- Alexandra C Brignall
- Montreal Neurological Institute, Centre for Neuronal Survival, 3801 University, Room MP105, Montréal, QC, H3A 2B4, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Jean-François Cloutier
- Montreal Neurological Institute, Centre for Neuronal Survival, 3801 University, Room MP105, Montréal, QC, H3A 2B4, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada.
| |
Collapse
|
21
|
Nakano H, Iida Y, Suzuki M, Aoki M, Umemura M, Takahashi S, Takahashi Y. Activating transcription factor 5 (ATF5) is essential for the maturation and survival of mouse basal vomeronasal sensory neurons. Cell Tissue Res 2015; 363:621-33. [DOI: 10.1007/s00441-015-2283-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/25/2015] [Indexed: 12/11/2022]
|
22
|
Stowers L, Kuo TH. Mammalian pheromones: emerging properties and mechanisms of detection. Curr Opin Neurobiol 2015; 34:103-9. [PMID: 25747731 DOI: 10.1016/j.conb.2015.02.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 12/22/2022]
Abstract
The concept of mammalian pheromones was established decades before the discovery of any bioactive ligands. Therefore, their molecular identity, native sources, and the meaning of their detection has been largely speculative. There has been recent success in identifying a variety of candidate mouse pheromones and other specialized odors. These discoveries reveal that mammalian pheromones come in a variety of ligand types and they are detected by sensory neurons that are pre-set to promote an array of social and survival behaviors. Importantly, recent findings show that they activate molecularly diverse sensory neurons that differ from canonical odorant detectors. These novel sensory neurons hold future promise to unlock the mystery of how their detection is hardwired to generate behavior.
Collapse
Affiliation(s)
- Lisa Stowers
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Tsung-Han Kuo
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
23
|
Churcher AM, Hubbard PC, Marques JP, Canário AVM, Huertas M. Deep sequencing of the olfactory epithelium reveals specific chemosensory receptors are expressed at sexual maturity in the European eel Anguilla anguilla. Mol Ecol 2015; 24:822-34. [PMID: 25580852 DOI: 10.1111/mec.13065] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 12/19/2014] [Accepted: 12/30/2014] [Indexed: 01/02/2023]
Abstract
Vertebrate genomes encode a diversity of G protein-coupled receptor (GPCR) that belong to large gene families and are used by olfactory systems to detect chemical cues found in the environment. It is not clear however, if individual receptors from these large gene families have evolved roles that are specific to certain life stages. Here, we used deep sequencing to identify differentially expressed receptor transcripts in the olfactory epithelia (OE) of freshwater, seawater and sexually mature male eels (Anguilla anguilla). This species is particularly intriguing because of its complex life cycle, extreme long-distance migrations and early-branching position within the teleost phylogeny. In the A. anguillaOE, we identified full-length transcripts for 13, 112, 6 and 38 trace amine-associated receptors, odorant receptors (OR) and type I and type II vomeronasal receptors (V1R and V2R). Most of these receptors were expressed at similar levels at different life stages and a subset of OR and V2R-like transcripts was more abundant in sexually mature males suggesting that ORs and V2R-like genes are important for reproduction. We also identified a set of GPCR signal transduction genes that were differentially expressed indicating that eels make use of different GPCR signal transduction genes at different life stages. The finding that a diversity of chemosensory receptors is expressed in the olfactory epithelium and that a subset is differentially expressed suggests that most receptors belonging to large chemosensory gene families have functions that are important at multiple life stages, while a subset has evolved specific functions at different life stages.
Collapse
Affiliation(s)
- Allison M Churcher
- Centro de Ciências do Mar, Universidade do Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | | | | | | | | |
Collapse
|
24
|
Pérez-Gómez A, Stein B, Leinders-Zufall T, Chamero P. Signaling mechanisms and behavioral function of the mouse basal vomeronasal neuroepithelium. Front Neuroanat 2014; 8:135. [PMID: 25505388 PMCID: PMC4244706 DOI: 10.3389/fnana.2014.00135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/03/2014] [Indexed: 01/20/2023] Open
Abstract
The vomeronasal organ (VNO) is a sensory organ that is found in most terrestrial vertebrates and that is principally implicated in the detection of pheromones. The VNO contains specialized sensory neurons organized in a pseudostratified neuroepithelium that recognize chemical signals involved in initiating innate behavioral responses. In rodents, the VNO neuroepithelium is segregated into two distinct zones, apical and basal. The molecular mechanisms involved in ligand detection by apical and basal VNO sensory neurons differ extensively. These two VNO subsystems express different subfamilies of vomeronasal receptors and signaling molecules, detect distinct chemosignals, and project to separate regions of the accessory olfactory bulb (AOB). The roles that these olfactory subdivisions play in the control of specific olfactory-mediated behaviors are largely unclear. However, analysis of mutant mouse lines for signal transduction components together with identification of defined chemosensory ligands has revealed a fundamental role of the basal part of the mouse VNO in mediating a wide range of instinctive behaviors, such as aggression, predator avoidance, and sexual attraction. Here we will compare the divergent functions and synergies between the olfactory subsystems and consider new insights in how higher neural circuits are defined for the initiation of instinctive behaviors.
Collapse
Affiliation(s)
- Anabel Pérez-Gómez
- Department of Physiology, University of Saarland School of Medicine Homburg, Saarland, Germany
| | - Benjamin Stein
- Department of Physiology, University of Saarland School of Medicine Homburg, Saarland, Germany
| | - Trese Leinders-Zufall
- Department of Physiology, University of Saarland School of Medicine Homburg, Saarland, Germany
| | - Pablo Chamero
- Department of Physiology, University of Saarland School of Medicine Homburg, Saarland, Germany
| |
Collapse
|
25
|
Cavaliere RM, Ghirardi F, Tirindelli R. Lacrimal gland removal impairs sexual behavior in mice. Front Neuroanat 2014; 8:101. [PMID: 25309342 PMCID: PMC4174856 DOI: 10.3389/fnana.2014.00101] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/05/2014] [Indexed: 11/13/2022] Open
Abstract
Exocrine gland-secreting peptides (ESPs) are a protein family involved in the pheromonal communication of rodents. ESP1 is a lacrimal peptide synthesized by the extraorbital glands of males of specific mouse strains that modulates the sexual behavior in females. Reportedly, BALB/c males, that produce high level of ESP1 in the tear fluid, were shown to enhance the lordosis behavior in C57BL/6 females during mating. In contrast, C57BL/6 and ICR males, both unable to express ESP1, failed to modulate this sexual behavior. Nonetheless, ICR males did become competent to enhance lordosis behavior in C57BL/6 females providing these were pre-exposed to ESP1. To exclude any strain differences, here, we investigated the pheromonal role of the extraorbital glands and indirectly of ESP1 in animals of the same strain. This was performed by applying the lordosis experimental paradigm in BALB/c mice before and after the surgical removal of these glands in males. The excision of the extraorbital glands reduced but did not abolish the production of ESP1 in the lacrimal fluid of BALB/c mice. An immunological analysis on soluble extracts of the glands that drain into the conjunctival sac revealed that the intraorbital glands (ILGs) are also responsible for the production of ESP1. The removal of both the extra and ILGs completely eliminated the tear secretion of ESP1. Extraorbital gland-deficient BALB/c mice were still able to induce lordosis behavior in sexually receptive females. In contrast, males with the removal of both the extra and ILGs failed to enhance lordosis behavior in females. Unexpectedly, C57BL/6 males did improve this sexual performance in BALB/c females. However, an analysis of the tear fluid of C57BL/6 males revealed low but detectable levels of ESP1. Overall, our study highlights the relevance of the orbital glands in modulating reproductive behavior and the sensitivity of the vomeronasal system to detect trace amount of ESP1.
Collapse
|
26
|
Oboti L, Pérez-Gómez A, Keller M, Jacobi E, Birnbaumer L, Leinders-Zufall T, Zufall F, Chamero P. A wide range of pheromone-stimulated sexual and reproductive behaviors in female mice depend on G protein Gαo. BMC Biol 2014. [PMID: 24886577 DOI: 10.1186/1741‐7007‐12‐31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Optimal reproductive fitness is essential for the biological success and survival of species. The vomeronasal organ is strongly implicated in the display of sexual and reproductive behaviors in female mice, yet the roles that apical and basal vomeronasal neuron populations play in controlling these gender-specific behaviors remain largely unclear. RESULTS To dissect the neural pathways underlying these functions, we genetically inactivated the basal vomeronasal organ layer using conditional, cell-specific ablation of the G protein Gαo. Female mice mutant for Gαo show severe alterations in sexual and reproductive behaviors, timing of puberty onset, and estrous cycle. These mutant mice are insensitive to reproductive facilitation stimulated by male pheromones that accelerate puberty and induce ovulation. Gαo-mutant females exhibit a striking reduction in sexual receptivity or lordosis behavior to males, but gender discrimination seems to be intact. These mice also show a loss in male scent preference, which requires a learned association for volatile olfactory signals with other nonvolatile ownership signals that are contained in the high molecular weight fraction of male urine. Thus, Gαo impacts on both instinctive and learned social responses to pheromones. CONCLUSIONS These results highlight that sensory neurons of the Gαo-expressing vomeronasal subsystem, together with the receptors they express and the molecular cues they detect, control a wide range of fundamental mating and reproductive behaviors in female mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Frank Zufall
- Department of Physiology, University of Saarland School of Medicine, 66421 Homburg, Germany.
| | | |
Collapse
|
27
|
Oboti L, Pérez-Gómez A, Keller M, Jacobi E, Birnbaumer L, Leinders-Zufall T, Zufall F, Chamero P. A wide range of pheromone-stimulated sexual and reproductive behaviors in female mice depend on G protein Gαo. BMC Biol 2014; 12:31. [PMID: 24886577 PMCID: PMC4038847 DOI: 10.1186/1741-7007-12-31] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/25/2014] [Indexed: 01/03/2023] Open
Abstract
Background Optimal reproductive fitness is essential for the biological success and survival of species. The vomeronasal organ is strongly implicated in the display of sexual and reproductive behaviors in female mice, yet the roles that apical and basal vomeronasal neuron populations play in controlling these gender-specific behaviors remain largely unclear. Results To dissect the neural pathways underlying these functions, we genetically inactivated the basal vomeronasal organ layer using conditional, cell-specific ablation of the G protein Gαo. Female mice mutant for Gαo show severe alterations in sexual and reproductive behaviors, timing of puberty onset, and estrous cycle. These mutant mice are insensitive to reproductive facilitation stimulated by male pheromones that accelerate puberty and induce ovulation. Gαo-mutant females exhibit a striking reduction in sexual receptivity or lordosis behavior to males, but gender discrimination seems to be intact. These mice also show a loss in male scent preference, which requires a learned association for volatile olfactory signals with other nonvolatile ownership signals that are contained in the high molecular weight fraction of male urine. Thus, Gαo impacts on both instinctive and learned social responses to pheromones. Conclusions These results highlight that sensory neurons of the Gαo-expressing vomeronasal subsystem, together with the receptors they express and the molecular cues they detect, control a wide range of fundamental mating and reproductive behaviors in female mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Frank Zufall
- Department of Physiology, University of Saarland School of Medicine, 66421 Homburg, Germany.
| | | |
Collapse
|