1
|
Wu M, He W, Gong H, Dong L, Ding N, Zhang G, Wang J, Rong W. The spatial and temporal pattern of GPER/GPR30 reporter expression in the developing and mature forebrain of mice. Brain Res Bull 2025; 223:111276. [PMID: 40024397 DOI: 10.1016/j.brainresbull.2025.111276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Evidence suggest that estrogens play crucial roles in the regulation of neural development and function and the G protein-coupled estrogen receptor (GPER/GPR30) appears to be the predominant estrogen receptor in the brain. However, the distribution and functions of GPER in the developing and mature brain are not fully understood. The current study aimed to characterize the expression of GPER in the forebrain, using Gper gene reporter mice combined with fluorescent in situ hybridization (FISH/RNAscope) and immunohistochemistry (IHC). Two lines of Gper reporter mice were constructed by crossing the Gper-cre mice with Ai14(RCL-tdT)-D or R26-ZsGreen mice, which showed identical spatial distributions of the reporters in adult brain. In the forebrain, neurons, protoplasmic astrocytes, mural cells and ependymal cells of third ventricle, were found to express Gper reporters. GPER-expressing neurons were particularly enriched in the olfactory system and the salience network, including posteromedial nucleus of the cortical amygdala (PmCo), entorhinal cortex, insula cortex, prefrontal cortex and dentate gyrus of the hippocampus. RNAscope and neural tracing showed GPER-expressing cortical neurons were long-range excitatory pyramidal neurons. GPER-expressing astrocytes represented a minor population (<10 %) of astrocytes and were found to be closely associated with neurovascular units. GPER-expressing mural cells were not labelled by the common pericyte marker PDGFRβ. In the critical period of neural development (P1-P10), GPER expression appeared to be intimately associated with neurogenesis, proliferation and migration in the olfactory system and the salience network. Collectively, the spatial and temporal pattern of GPER/GPR30 expression in the forebrain implied it might play important roles regulating the development and functions of the olfactory system, the salience network and the cerebral vessels.
Collapse
Affiliation(s)
- Meimei Wu
- Department of Gastroenterology, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenxin He
- School of Basic Medical Science and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan 750004, China
| | - Huashan Gong
- Department of Gastroenterology, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li Dong
- Department of Gastroenterology, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Na Ding
- School of Basic Medical Science and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan 750004, China
| | - Guohua Zhang
- Department of Gastroenterology, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Wang
- Department of Gastroenterology, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Weifang Rong
- Department of Gastroenterology, Songjiang Research Institute, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; School of Basic Medical Science and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
2
|
Abstract
Historically, the human sense of smell has been regarded as the odd stepchild of the senses, especially compared to the sensory bravado of seeing, touching, and hearing. The idea that the human olfaction has little to contribute to our experience of the world is commonplace, though with the emergence of COVID-19 there has rather been a sea change in this understanding. An ever increasing body of work has convincingly highlighted the keen capabilities of the human nose and the sophistication of the human olfactory system. Here, we provide a concise overview of the neuroscience of human olfaction spanning the last 10-15 years, with focus on the peripheral and central mechanisms that underlie how odor information is processed, packaged, parceled, predicted, and perturbed to serve odor-guided behaviors. We conclude by offering some guideposts for harnessing the next decade of olfactory research in all its shapes and forms.
Collapse
Affiliation(s)
| | - Jay A Gottfried
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
- Department of Psychology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
From In Vitro Data to In Vivo Interspecies Danger Communication: A Study of Chemosensing via the Mouse Grueneberg Ganglion. Animals (Basel) 2022; 12:ani12030356. [PMID: 35158677 PMCID: PMC8833560 DOI: 10.3390/ani12030356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary The mouse olfactory system is essential for danger detection with a critical role in the Grueneberg ganglion subsystem. This organ, which is localized at the tip of the nose, is implicated in the recognition of kairomones, or chemical cues released by predators which allow interspecies communication. These kairomones, which are present in the secretions of predators, will induce fear-related behaviours in mice. It is not yet known how the Grueneberg ganglion neurons can detect these molecules; however, three specific bitter taste receptors, known as TAS2Rs, that are present in the Grueneberg ganglion play a role in this detection. Here, using in vitro, ex vivo and in vivo experimental approaches, we identified two novel and potent sources of kairomones that are recognized by the mouse Grueneberg ganglion neurons, namely the biological secretions from the raccoon (Procyon lotor) and the skunk (Mephitis mephitis). Abstract In the wild, mice have developed survival strategies to detect volatile cues that warn them of potential danger. Specific olfactory neurons found in the Grueneberg ganglion olfactory subsystem can detect alarm pheromones emitted by stressed conspecifics, as well as kairomones involuntarily released by their predators. These volatile chemical cues allow intra- and interspecies communication of danger, respectively. Alarm pheromones, kairomones and bitter taste ligands share a common chemical motif containing sulfur or nitrogen. Interestingly, three specific bitter taste receptors (TAS2Rs) have been found in the Grueneberg ganglion neurons that are implicated in danger signalling pathways. We have recently developed a TAS2R–expressing heterologous system that mimics the Grueneberg ganglion neuron responses after kairomone stimulation. Here, we demonstrated by in vitro, ex vivo and in vivo experiments that the biological secretions from the raccoon (Procyon lotor) and the skunk (Mephitis mephitis) were acting as potent sources of kairomones. They activated the Grueneberg ganglion neurons and induced fear-related behaviours in mice. Identification of new sources of semiochemicals is a first step towards an understanding of the interspecies danger communication that takes place in the Grueneberg ganglion.
Collapse
|
4
|
Pardasani M, Marathe SD, Purnapatre MM, Dalvi U, Abraham NM. Multimodal learning of pheromone locations. FASEB J 2021; 35:e21836. [PMID: 34407246 PMCID: PMC7611819 DOI: 10.1096/fj.202100167r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/28/2021] [Accepted: 07/20/2021] [Indexed: 11/11/2022]
Abstract
Memorizing pheromonal locations is critical for many mammalian species as it involves finding mates and avoiding competitors. In rodents, pheromonal information is perceived by the main and accessory olfactory systems. However, the role of somatosensation in context-dependent learning and memorizing of pheromone locations remains unexplored. We addressed this problem by training female mice on a multimodal task to locate pheromones by sampling volatiles emanating from male urine through the orifices of varying dimensions or shapes that are sensed by their vibrissae. In this novel pheromone location assay, female mice’ preference toward male urine scent decayed over time when they were permitted to explore pheromones vs neutral stimuli, water. On training them for the associations involving olfactory and whisker systems, it was established that they were able to memorize the location of opposite sex pheromones, when tested 15 days later. This memory was not formed either when the somatosensory inputs through whisker pad were blocked or when the pheromonal cues were replaced with that of same sex. The association between olfactory and somatosensory systems was further confirmed by the enhanced expression of the activity-regulated cytoskeleton protein. Furthermore, the activation of main olfactory bulb circuitry by pheromone volatiles did not cause any modulation in learning and memorizing non-pheromonal volatiles. Our study thus provides the evidence for associations formed between different sensory modalities facilitating the long-term memory formation relevant to social and reproductive behaviors.
Collapse
Affiliation(s)
- Meenakshi Pardasani
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| | - Shruti D Marathe
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| | - Maitreyee Mandar Purnapatre
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India.,Institute of Bioinformatics & Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Urvashi Dalvi
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India.,Institute of Bioinformatics & Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Nixon M Abraham
- Laboratory of Neural Circuits and Behaviour (LNCB), Department of Biology, Indian Institute of Science Education and Research (IISER), Pune, India
| |
Collapse
|
5
|
Kolter JF, Hildenbrand MF, Popp S, Nauroth S, Bankmann J, Rother L, Waider J, Deckert J, Asan E, Jakob PM, Lesch KP, Schmitt-Böhrer A. Serotonin transporter genotype modulates resting state and predator stress-induced amygdala perfusion in mice in a sex-dependent manner. PLoS One 2021; 16:e0247311. [PMID: 33606835 PMCID: PMC7895400 DOI: 10.1371/journal.pone.0247311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022] Open
Abstract
The serotonin transporter (5-HTT) is a key molecule of serotoninergic neurotransmission and target of many anxiolytics and antidepressants. In humans, 5-HTT gene variants resulting in lower expression levels are associated with behavioral traits of anxiety. Furthermore, functional magnetic resonance imaging (fMRI) studies reported increased cerebral blood flow (CBF) during resting state (RS) and amygdala hyperreactivity. 5-HTT deficient mice as an established animal model for anxiety disorders seem to be well suited for investigating amygdala (re-)activity in an fMRI study. We investigated wildtype (5-HTT+/+), heterozygous (5-HTT+/-), and homozygous 5-HTT-knockout mice (5-HTT-/-) of both sexes in an ultra-high-field 17.6 Tesla magnetic resonance scanner. CBF was measured with continuous arterial spin labeling during RS, stimulation state (SS; with odor of rats as aversive stimulus), and post-stimulation state (PS). Subsequently, post mortem c-Fos immunohistochemistry elucidated neural activation on cellular level. The results showed that in reaction to the aversive odor CBF in total brain and amygdala of all mice significantly increased. In male 5-HTT+/+ mice amygdala RS CBF levels were found to be significantly lower than in 5-HTT+/- mice. From RS to SS 5-HTT+/+ amygdala perfusion significantly increased compared to both 5-HTT+/- and 5-HTT-/- mice. Perfusion level changes of male mice correlated with the density of c-Fos-immunoreactive cells in the amygdaloid nuclei. In female mice the perfusion was not modulated by the 5-Htt-genotype, but by estrous cycle stages. We conclude that amygdala reactivity is modulated by the 5-Htt genotype in males. In females, gonadal hormones have an impact which might have obscured genotype effects. Furthermore, our results demonstrate experimental support for the tonic model of 5-HTTLPR function.
Collapse
Affiliation(s)
- Jann F. Kolter
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Markus F. Hildenbrand
- Department of Magnetic Resonance and X-Ray Imaging, Fraunhofer Development Center X-Ray Technology, Wuerzburg, Germany
| | - Sandy Popp
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Stephan Nauroth
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Julian Bankmann
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Lisa Rother
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Jonas Waider
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Jürgen Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Esther Asan
- Institute of Anatomy and Cell Biology, University of Wuerzburg, Wuerzburg, Germany
| | - Peter M. Jakob
- Department of Experimental Physics 5, University of Wuerzburg, Wuerzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Angelika Schmitt-Böhrer
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
6
|
Braubach O, Croll RP. The glomerular network of the zebrafish olfactory bulb. Cell Tissue Res 2021; 383:255-271. [PMID: 33484356 DOI: 10.1007/s00441-020-03394-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/25/2020] [Indexed: 10/22/2022]
Abstract
Each zebrafish olfactory bulb contains ~ 140 glomeruli that are distinguishable based on size, location, neurochemistry and function. Here we examine the mitral cell innervation of differently sized glomeruli in adult zebrafish. Type 1 glomeruli had diameters of 80.9 ± 8.1 μm and were innervated by 5.9 ± 0.9 mitral cells. The Type 1 mediodorsal glomeruli (mdG) were innervated by both uniglomerular (innervating only single glomeruli) and multiglomerular mitral cells (innervating two or more glomeruli). In contrast, the Type 1 ventroposterior (vpG) and lateral glomeruli (lG) were only innervated by uniglomerular mitral cells. Type 2 ventral glomeruli were 46 ± 5.1 μm in diameter and were innervated by 3.3 ± 0.2 mitral cells. Type 2 ventromedial glomeruli (vmG) were innervated exclusively by uniglomerular mitral cells. Type 3 glomeruli had diameters of 17 ± 2.5 μm and were innervated by 1.1 ± 0.6 multiglomerular mitral cells each. Finally, Type 4 glomeruli were small, with average diameters of 4.8 ± 3.9 μm and were restricted to the lateral plexus. These glomeruli were innervated mainly by multiglomerular mitral cells with extensively branching dendrites. This study provides the first specific associations between uni- and multiglomerular mitral cells with known zebrafish glomeruli. Our results suggest that glomeruli are distinguishable based on their postsynaptic compartment and that distinct input-output computations occur in different types of zebrafish glomeruli.
Collapse
Affiliation(s)
- Oliver Braubach
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, B3H4R2, Canada.
| | - Roger P Croll
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, B3H4R2, Canada
| |
Collapse
|
7
|
Levy DR, Sofer Y, Brumfeld V, Zilkha N, Kimchi T. The Nasopalatine Ducts Are Required for Proper Pheromone Signaling in Mice. Front Neurosci 2020; 14:585323. [PMID: 33328853 PMCID: PMC7710809 DOI: 10.3389/fnins.2020.585323] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/07/2020] [Indexed: 11/25/2022] Open
Abstract
The vomeronasal organ (VNO) specializes in detection of chemosignals, mainly pheromones, which control social communication and reproduction in many mammals. These pheromones must solubilize with nasal fluids before entering the VNO, and it was suggested that they are delivered to and cleared from the VNO by active pumping. Yet, the details of this pheromone delivery process are unclear. In this study, we first constructed a high-resolution 3D morphological image of the whole adult mouse snout, by using ultra-high-resolution micro-CT. We identified a net of micro tunnels starting from the nostrils and extending around and through the VNO. These micro tunnels connect the nasal cavity with the VNO and the oral cavity via the nasopalatine ducts (NPD). Other micro tunnels connect the nasal cavity to the main olfactory epithelium. We next demonstrated that physical obstruction of the NPD severely impairs the clearance of dissolved compounds from the VNO lumen. Moreover, we found that mice with blocked NPD display alterations in chemosignaling-evoked neuronal activation in brain regions associated with the vomeronasal system. Finally, NPD-blocked male mice exhibit reduced preference for female chemosignals, and impaired social interaction behavior. Taken together, our findings indicate that the NPD in mice are connected to both the nasal and oral cavity, serving an essential role in regulating the flow of soluble chemosignals through the VNO, and are required for proper pheromone-mediated social communication.
Collapse
Affiliation(s)
- Dana Rubi Levy
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Yizhak Sofer
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Vlad Brumfeld
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Noga Zilkha
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Kimchi
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
8
|
Pandolfi EC, Hoffmann HM, Schoeller EL, Gorman MR, Mellon PL. Haploinsufficiency of SIX3 Abolishes Male Reproductive Behavior Through Disrupted Olfactory Development, and Impairs Female Fertility Through Disrupted GnRH Neuron Migration. Mol Neurobiol 2018; 55:8709-8727. [PMID: 29589282 PMCID: PMC6156938 DOI: 10.1007/s12035-018-1013-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/16/2018] [Indexed: 12/17/2022]
Abstract
Mating behavior in males and females is dependent on olfactory cues processed through both the main olfactory epithelium (MOE) and the vomeronasal organ (VNO). Signaling through the MOE is critical for the initiation of male mating behavior, and the loss of MOE signaling severely compromises this comportment. Here, we demonstrate that dosage of the homeodomain gene Six3 affects the degree of development of MOE but not the VNO. Anomalous MOE development in Six3 heterozygote mice leads to hyposmia, specifically disrupting male mounting behavior by impairing detection of volatile female estrus pheromones. Six3 is highly expressed in the MOE, main olfactory bulb (MOB), and hypothalamus; all regions essential in the proper migration of the gonadotropin-releasing hormone (GnRH) neurons, a key reproductive neuronal population that migrates along olfactory axons from the developing nose into the brain. Interestingly, we find that the reduction in Six3 expression in Six3 heterozygote mice compromises development of the MOE and MOB, resulting in mis-migration of GnRH neurons due to improper olfactory axon targeting. This reduction in the hypothalamic GnRH neuron population, by 45% in adulthood, leads to female subfertility, but does not impact male hormone levels, suggesting that male infertility is not related to GnRH neuron numbers, but exclusively linked to abnormal olfaction. We here determine that Six3 is haploinsufficient for MOE development, GnRH neuron migration, and fertility, and represents a novel candidate gene for Kallmann syndrome, a form of inherited infertility.
Collapse
Affiliation(s)
- Erica C Pandolfi
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Hanne M Hoffmann
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Erica L Schoeller
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael R Gorman
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Psychology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Pamela L Mellon
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA.
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
9
|
The Neural Mechanisms of Sexually Dimorphic Aggressive Behaviors. Trends Genet 2018; 34:755-776. [PMID: 30173869 DOI: 10.1016/j.tig.2018.07.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/16/2018] [Accepted: 07/05/2018] [Indexed: 10/28/2022]
Abstract
Aggression is a fundamental social behavior that is essential for competing for resources and protecting oneself and families in both males and females. As a result of natural selection, aggression is often displayed differentially between the sexes, typically at a higher level in males than females. Here, we highlight the behavioral differences between male and female aggression in rodents. We further outline the aggression circuits in males and females, and compare their differences at each circuit node. Lastly, we summarize our current understanding regarding the generation of sexually dimorphic aggression circuits during development and their maintenance during adulthood. In both cases, gonadal steroid hormones appear to play crucial roles in differentiating the circuits by impacting on the survival, morphology, and intrinsic properties of relevant cells. Many other factors, such as environment and experience, may also contribute to sex differences in aggression and remain to be investigated in future studies.
Collapse
|
10
|
Harmeier A, Meyer CA, Staempfli A, Casagrande F, Petrinovic MM, Zhang YP, Künnecke B, Iglesias A, Höner OP, Hoener MC. How Female Mice Attract Males: A Urinary Volatile Amine Activates a Trace Amine-Associated Receptor That Induces Male Sexual Interest. Front Pharmacol 2018; 9:924. [PMID: 30158871 PMCID: PMC6104183 DOI: 10.3389/fphar.2018.00924] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/26/2018] [Indexed: 11/27/2022] Open
Abstract
Individuals of many species rely on odors to communicate, find breeding partners, locate resources and sense dangers. In vertebrates, odorants are detected by chemosensory receptors of the olfactory system. One class of these receptors, the trace amine-associated receptors (TAARs), was recently suggested to mediate male sexual interest and mate choice. Here we tested this hypothesis in mice by generating a cluster deletion mouse (Taar2-9−/−) lacking all TAARs expressed in the olfactory epithelium, and evaluating transduction pathways from odorants to TAARs, neural activity and behaviors reflecting sexual interest. We found that a urinary volatile amine, isobutylamine (IBA), was a potent ligand for TAAR3 (but not TAAR1, 4, 5, and 6). When males were exposed to IBA, brain regions associated with sexual behaviors were less active in Taar2-9−/− than in wild type males. Accordingly, Taar2-9−/− males spent less time sniffing both the urine of females and pure IBA than wild type males. This is the first demonstration of a comprehensive transduction pathway linking odorants to TAARs and male sexual interest. Interestingly, the concentration of IBA in female urine varied across the estrus cycle with a peak during estrus. This variation in IBA concentration may represent a simple olfactory cue for males to recognize receptive females. Our results are consistent with the hypothesis that IBA and TAARs play an important role in the recognition of breeding partners and mate choice.
Collapse
Affiliation(s)
- Anja Harmeier
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Claas A Meyer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Andreas Staempfli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Fabio Casagrande
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Marija M Petrinovic
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.,Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Yan-Ping Zhang
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Basil Künnecke
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Antonio Iglesias
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Oliver P Höner
- Department of Evolutionary Ecology, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Marius C Hoener
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.,Department of Neurosymptomatic Domains, Neuroscience, Ophthalmology and Rare Diseases Discovery and Translational Area, Roche Pharma Research and Early Development pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
11
|
Silvotti L, Cavaliere RM, Belletti S, Tirindelli R. In-vivo activation of vomeronasal neurons shows adaptive responses to pheromonal stimuli. Sci Rep 2018; 8:8490. [PMID: 29855521 PMCID: PMC5981476 DOI: 10.1038/s41598-018-26831-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/18/2018] [Indexed: 01/29/2023] Open
Abstract
In most mammals, the vomeronasal system has a pivotal role in mediating socio-sexual behaviours. The vomeronasal organ senses pheromones through the activation of specific receptors. Pheromone binding to cognate receptors activates Ca-influx via the gating of a cation channel that generates membrane depolarisation. The ex-vivo activation of vomeronasal neurons (VSNs) by pheromonal stimuli has been largely investigated by electrophysiological and imaging techniques; however, few studies have been carried out to determine the physiological responses of VSNs, in-vivo. By tracking the phosphorylation of S6 ribosomal protein as a marker of neuronal activity, we show that S6 becomes phosphorylated (pS6) in mouse VSNs stimulated by intraspecific and heterospecific pheromonal cues. We observed that female scent induces pS6 immunoreactivity in the apical VSNs of male vomeronasal epithelium, whereas male cues stimulate S6 phosphorylation in both the basal and apical VSNs of females. We also show that this dimorphic pattern of pS6 immunoreactivity is reproduced when heterospecific stimuli are used. Moreover, we found that a consistent proportion of VSNs is activated by both heterospecific and intraspecific pheromones. Additionally, we have evidence of adaptive responses to S6 phosphorylation when stimulation with cues of the same and opposite sex and of different species is sustained.
Collapse
Affiliation(s)
- Lucia Silvotti
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Via Volturno, 39, 43125, Parma, Italy
| | - Rosa Maria Cavaliere
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Via Volturno, 39, 43125, Parma, Italy
| | - Silvana Belletti
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Via Volturno, 39, 43125, Parma, Italy
| | - Roberto Tirindelli
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Via Volturno, 39, 43125, Parma, Italy.
| |
Collapse
|
12
|
Loss of Kirrel family members alters glomerular structure and synapse numbers in the accessory olfactory bulb. Brain Struct Funct 2017; 223:307-319. [DOI: 10.1007/s00429-017-1485-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/24/2017] [Indexed: 10/19/2022]
|
13
|
Korsak LIT, Shepard KA, Akins MR. Cell type-dependent axonal localization of translational regulators and mRNA in mouse peripheral olfactory neurons. J Comp Neurol 2017; 525:2202-2215. [PMID: 28266018 DOI: 10.1002/cne.24199] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 01/01/2023]
Abstract
Local protein synthesis in mature axons may play a role in synaptic plasticity, axonal arborization, or functional diversity of the circuit. To gain insight into this question, we investigated the axonal localization of translational regulators and associated mRNAs in five parallel olfactory circuits, four in the main olfactory bulb and one in the accessory olfactory bulb. Axons in all four main olfactory bulb circuits exhibited axonal localization of Fragile X granules (FXGs), structures that comprise ribosomes, mRNA, and RNA binding proteins including Fragile X mental retardation protein (FMRP) and the related protein FXR2P. In contrast, FXGs were not seen in axons innervating the accessory olfactory bulb. Similarly, axons innervating the main olfactory bulb, but not the accessory olfactory bulb, contained the FXG-associated mRNA Omp (olfactory marker protein). This differential localization was not explained by circuit-dependent differences in expression of FXG components or Omp, suggesting that other factors must regulate their axonal transport. The specificity of this transport was highlighted by the absence from olfactory axons of the calmodulin transcript Calm1, which is highly expressed in peripheral olfactory neurons at levels equivalent to Omp. Regulation of axonal translation by FMRP may shape the structure and function of the axonal arbor in mature sensory neurons in the main olfactory system but not in the accessory olfactory system.
Collapse
Affiliation(s)
- Lulu I T Korsak
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, 19104
| | | | - Michael R Akins
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, 19104.,Department of Neurobiology and Anatomy, Drexel University, Philadelphia, Pennsylvania, 19104
| |
Collapse
|
14
|
Abstract
In this review, I discuss current knowledge and outstanding questions on the neuromodulators that influence aggressive behavior of the fruit fly Drosophila melanogaster. I first present evidence that Drosophila exchange information during an agonistic interaction and choose appropriate actions based on this information. I then discuss the influence of several biogenic amines and neuropeptides on aggressive behavior. One striking characteristic of neuromodulation is that it can configure a neural circuit dynamically, enabling one circuit to generate multiple outcomes. I suggest a consensus effect of each neuromodulatory molecule on Drosophila aggression, as well as effects of receptor proteins where relevant data are available. Lastly, I consider neuromodulation in the context of strategic action choices during agonistic interactions. Genetic components of neuromodulatory systems are highly conserved across animals, suggesting that molecular and cellular mechanisms controlling Drosophila aggression can shed light on neural principles governing action choice during social interactions.
Collapse
Affiliation(s)
- Kenta Asahina
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037;
| |
Collapse
|
15
|
Abstract
The sense of smell is mediated by GPCRs in the odorant receptor (OR) family. Greer et al. report a new family of odor detectors, MS4As, that have similar cellular localization and chemodetection ability as ORs but are not GPCRs and follow a strikingly different logic of odor coding at the periphery.
Collapse
|
16
|
Silva BA, Gross CT, Gräff J. The neural circuits of innate fear: detection, integration, action, and memorization. ACTA ACUST UNITED AC 2016; 23:544-55. [PMID: 27634145 PMCID: PMC5026211 DOI: 10.1101/lm.042812.116] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/19/2016] [Indexed: 12/26/2022]
Abstract
How fear is represented in the brain has generated a lot of research attention, not only because fear increases the chances for survival when appropriately expressed but also because it can lead to anxiety and stress-related disorders when inadequately processed. In this review, we summarize recent progress in the understanding of the neural circuits processing innate fear in rodents. We propose that these circuits are contained within three main functional units in the brain: a detection unit, responsible for gathering sensory information signaling the presence of a threat; an integration unit, responsible for incorporating the various sensory information and recruiting downstream effectors; and an output unit, in charge of initiating appropriate bodily and behavioral responses to the threatful stimulus. In parallel, the experience of innate fear also instructs a learning process leading to the memorization of the fearful event. Interestingly, while the detection, integration, and output units processing acute fear responses to different threats tend to be harbored in distinct brain circuits, memory encoding of these threats seems to rely on a shared learning system.
Collapse
Affiliation(s)
- Bianca A Silva
- Laboratory of Neuroepigenetics, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale Lausanne, CH-1015 Lausanne, Switzerland
| | - Cornelius T Gross
- Mouse Biology Unit, European Molecular Biology Laboratory (EMBL), 00015 Monterotondo, Italy
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
17
|
Stempel H, Jung M, Pérez-Gómez A, Leinders-Zufall T, Zufall F, Bufe B. Strain-specific Loss of Formyl Peptide Receptor 3 in the Murine Vomeronasal and Immune Systems. J Biol Chem 2016; 291:9762-75. [PMID: 26957543 DOI: 10.1074/jbc.m116.714493] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 12/30/2022] Open
Abstract
Formyl peptide receptor 3 (Fpr3, also known as Fpr-rs1) is a G protein-coupled receptor expressed in subsets of sensory neurons of the mouse vomeronasal organ, an olfactory substructure essential for social recognition. Fpr3 has been implicated in the sensing of infection-associated olfactory cues, but its expression pattern and function are incompletely understood. To facilitate visualization of Fpr3-expressing cells, we generated and validated two new anti-Fpr3 antibodies enabling us to analyze acute Fpr3 protein expression. Fpr3 is not only expressed in murine vomeronasal sensory neurons but also in bone marrow cells, the primary source for immune cell renewal, and in mature neutrophils. Consistent with the notion that Fpr3 functions as a pathogen sensor, Fpr3 expression in the immune system is up-regulated after stimulation with a bacterial endotoxin (lipopolysaccharide). These results strongly support a dual role for Fpr3 in both vomeronasal sensory neurons and immune cells. We also identify a large panel of mouse strains with severely altered expression and function of Fpr3, thus establishing the existence of natural Fpr3 knock-out strains. We attribute distinct Fpr3 expression in these strains to the presence or absence of a 12-nucleotide in-frame deletion (Fpr3Δ424-435). In vitro calcium imaging and immunofluorescence analyses demonstrate that the lack of four amino acids leads to an unstable, truncated, and non-functional receptor protein. The genome of at least 19 strains encodes a non-functional Fpr3 variant, whereas at least 13 other strains express an intact receptor. These results provide a foundation for understanding the in vivo function of Fpr3.
Collapse
Affiliation(s)
- Hendrik Stempel
- From the Center for Integrative Physiology and Molecular Medicine and
| | - Martin Jung
- the Department of Biochemistry, Saarland University School of Medicine, 66421 Homburg, Germany
| | | | | | - Frank Zufall
- From the Center for Integrative Physiology and Molecular Medicine and
| | - Bernd Bufe
- From the Center for Integrative Physiology and Molecular Medicine and
| |
Collapse
|
18
|
Hashikawa K, Hashikawa Y, Falkner A, Lin D. The neural circuits of mating and fighting in male mice. Curr Opin Neurobiol 2016; 38:27-37. [PMID: 26849838 DOI: 10.1016/j.conb.2016.01.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/09/2016] [Indexed: 01/02/2023]
Abstract
Tinbergen proposed that instinctive behaviors can be divided into appetitive and consummatory phases. During mating and aggression, the appetitive phase contains various actions to bring an animal to a social target and the consummatory phase allows stereotyped actions to take place. Here, we summarize recent advances in elucidating the neural circuits underlying the appetitive and consummatory phases of sexual and aggressive behaviors with a focus on male mice. We outline the role of the main olfactory inputs in the initiation of social approach; the engagement of the accessory olfactory system during social investigation, and the role of the hypothalamus and its downstream pathways in orchestrating social behaviors through a suite of motor actions.
Collapse
Affiliation(s)
- Koichi Hashikawa
- Institute of Neuroscience, New York University School of Medicine, New York, NY 10016, USA
| | - Yoshiko Hashikawa
- Institute of Neuroscience, New York University School of Medicine, New York, NY 10016, USA
| | - Annegret Falkner
- Institute of Neuroscience, New York University School of Medicine, New York, NY 10016, USA
| | - Dayu Lin
- Institute of Neuroscience, New York University School of Medicine, New York, NY 10016, USA; Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
19
|
Ben-Shaul Y. Extracting Social Information from Chemosensory Cues: Consideration of Several Scenarios and Their Functional Implications. Front Neurosci 2015; 9:439. [PMID: 26635515 PMCID: PMC4653286 DOI: 10.3389/fnins.2015.00439] [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] [Received: 08/31/2015] [Accepted: 10/30/2015] [Indexed: 11/16/2022] Open
Abstract
Across all sensory modalities, stimuli can vary along multiple dimensions. Efficient extraction of information requires sensitivity to those stimulus dimensions that provide behaviorally relevant information. To derive social information from chemosensory cues, sensory systems must embed information about the relationships between behaviorally relevant traits of individuals and the distributions of the chemical cues that are informative about these traits. In simple cases, the mere presence of one particular compound is sufficient to guide appropriate behavior. However, more generally, chemosensory information is conveyed via relative levels of multiple chemical cues, in non-trivial ways. The computations and networks needed to derive information from multi-molecule stimuli are distinct from those required by single molecule cues. Our current knowledge about how socially relevant information is encoded by chemical blends, and how it is extracted by chemosensory systems is very limited. This manuscript explores several scenarios and the neuronal computations required to identify them.
Collapse
Affiliation(s)
- Yoram Ben-Shaul
- Department of Medical Neurobiology, Hebrew University Medical School Jerusalem, Israel
| |
Collapse
|
20
|
Rosen JB, Asok A, Chakraborty T. The smell of fear: innate threat of 2,5-dihydro-2,4,5-trimethylthiazoline, a single molecule component of a predator odor. Front Neurosci 2015; 9:292. [PMID: 26379483 PMCID: PMC4548190 DOI: 10.3389/fnins.2015.00292] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022] Open
Abstract
In the last several years, the importance of understanding what innate threat and fear is, in addition to learning of threat and fear, has become evident. Odors from predators are ecologically relevant stimuli used by prey animals as warnings for the presence of danger. Of importance, these odors are not necessarily noxious or painful, but they have innate threat-like properties. This review summarizes the progress made on the behavioral and neuroanatomical fundamentals of innate fear of the predator odor, 2,5-dihydro-2,4,5-trimethylthiazoline (TMT), a component of fox feces. TMT is one of several single molecule components of predator odors that have been isolated in the last several years. Isolation of these single molecules has allowed for rapid advances in delineating the behavioral constraints and selective neuroanatomical pathways of predator odor induced fear. In naïve mice and rats, TMT induces a number of fear and defensive behaviors, including robust freezing, indicating it is an innate threat stimulus. However, there are a number of behavioral constraints that we do not yet understand. Similarly, while some of the early olfactory sensory pathways for TMT-induced fear are being delineated, the pathways from olfactory systems to emotional and motor output regions are less well understood. This review will focus on what we know and what we still need to learn about the behavior and neuroanatomy of TMT-induced fear.
Collapse
Affiliation(s)
- Jeffrey B. Rosen
- Department of Psychological and Brain Sciences, University of DelawareNewark, DE, USA
| | | | | |
Collapse
|
21
|
Lewis L, Siju K, Aso Y, Friedrich A, Bulteel A, Rubin G, Grunwald Kadow I. A Higher Brain Circuit for Immediate Integration of Conflicting Sensory Information in Drosophila. Curr Biol 2015; 25:2203-14. [DOI: 10.1016/j.cub.2015.07.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/18/2015] [Accepted: 07/06/2015] [Indexed: 10/23/2022]
|
22
|
Root CM, Denny CA, Hen R, Axel R. The participation of cortical amygdala in innate, odour-driven behaviour. Nature 2014; 515:269-73. [PMID: 25383519 PMCID: PMC4231015 DOI: 10.1038/nature13897] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 09/26/2014] [Indexed: 11/18/2022]
Abstract
Innate behaviors are observed in naïve animals without prior learning or experience, suggesting that the neural circuits that mediate these behaviors are genetically determined and stereotyped. The neural circuits that convey olfactory information from the sense organ to the cortical and subcortical olfactory centers have been anatomically defined1-3 but the specific pathways responsible for innate responses to volatile odors have not been identified. We have devised genetic strategies that demonstrate that a stereotyped neural circuit that transmits information from the olfactory bulb to cortical amygdala is necessary for innate aversive and appetitive behaviors. Moreover, we have employed the promoter of the activity-dependent gene, arc, to express the photosensitive ion channel, channelrhodopsin, in neurons of the cortical amygdala activated by odors that elicit innate behaviors. Optical activation of these neurons leads to appropriate behaviors that recapitulate the responses to innate odors. These data indicate that the cortical amygdala plays a critical role in the generation of innate odor-driven behaviors but do not preclude the participation of cortical amygdala in learned olfactory behaviors.
Collapse
Affiliation(s)
- Cory M Root
- Department of Neuroscience and the Howard Hughes Medical Institute, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Christine A Denny
- 1] Department of Biological Sciences, New York State Psychiatric Institute, New York, New York 10032, USA [2] Department of Neuroscience and Psychiatry, New York State Psychiatric Institute, New York, New York 10032, USA [3] Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032, USA
| | - René Hen
- 1] Department of Neuroscience and Psychiatry, New York State Psychiatric Institute, New York, New York 10032, USA [2] Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York 10032, USA [3] Department of Pharmacology, Columbia University, New York State Psychiatric Institute, New York, New York 10032, USA
| | - Richard Axel
- Department of Neuroscience and the Howard Hughes Medical Institute, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| |
Collapse
|
23
|
|
24
|
|
25
|
Abstract
The olfactory (OR) and vomeronasal receptor (VR) repertoires are collectively encoded by 1700 genes and pseudogenes in the mouse genome. Most OR and VR genes were identified by comparative genomic techniques and therefore, in many of those cases, only their protein coding sequences are defined. Some also lack experimental support, due in part to the similarity between them and their monogenic, cell-specific expression in olfactory tissues. Here we use deep RNA sequencing, expression microarray and quantitative RT-PCR in both the vomeronasal organ and whole olfactory mucosa to quantify their full transcriptomes in multiple male and female mice. We find evidence of expression for all VR, and almost all OR genes that are annotated as functional in the reference genome, and use the data to generate over 1100 new, multi-exonic, significantly extended receptor gene annotations. We find that OR and VR genes are neither equally nor randomly expressed, but have reproducible distributions of abundance in both tissues. The olfactory transcriptomes are only minimally different between males and females, suggesting altered gene expression at the periphery is unlikely to underpin the striking sexual dimorphism in olfactory-mediated behavior. Finally, we present evidence that hundreds of novel, putatively protein-coding genes are expressed in these highly specialized olfactory tissues, and carry out a proof-of-principle validation. Taken together, these data provide a comprehensive, quantitative catalog of the genes that mediate olfactory perception and pheromone-evoked behavior at the periphery. The sense of smell in mice involves the detection of odors and pheromones by many hundreds of olfactory and vomeronasal receptors. The genes that encode these receptors account for around 5% of the whole gene catalog, but they are poorly understood because they are very similar to each other, and are thought to be turned on randomly in only a small number of cells. Here we use multiple gene expression technologies to curate and measure the activity of all the genes involved in the detection of odors and find evidence of many new ones. We show that most genes encoding olfactory and vomeronasal receptors have complex, multi-exonic structures that generate different isoforms. We find that some receptors are consistently more abundant in the nose than others, which suggests they are not turned on randomly. This may explain why mice are particularly sensitive to some odors, but less attuned to others. We find that overall males and females differ very little in gene expression, despite having altered behavioral responses to the same odors. Thus diversity in receptor expression can explain differences in odor sensitivity, but does not appear to dictate whether sex pheromones are differentially detected by males or females.
Collapse
|
26
|
Asahina K, Watanabe K, Duistermars BJ, Hoopfer E, González CR, Eyjólfsdóttir EA, Perona P, Anderson DJ. Tachykinin-expressing neurons control male-specific aggressive arousal in Drosophila. Cell 2014; 156:221-35. [PMID: 24439378 DOI: 10.1016/j.cell.2013.11.045] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 09/10/2013] [Accepted: 11/15/2013] [Indexed: 11/29/2022]
Abstract
Males of most species are more aggressive than females, but the neural mechanisms underlying this dimorphism are not clear. Here, we identify a neuron and a gene that control the higher level of aggression characteristic of Drosophila melanogaster males. Males, but not females, contain a small cluster of FruM(+) neurons that express the neuropeptide tachykinin (Tk). Activation and silencing of these neurons increased and decreased, respectively, intermale aggression without affecting male-female courtship behavior. Mutations in both Tk and a candidate receptor, Takr86C, suppressed the effect of neuronal activation, whereas overexpression of Tk potentiated it. Tk neuron activation overcame reduced aggressiveness caused by eliminating a variety of sensory or contextual cues, suggesting that it promotes aggressive arousal or motivation. Tachykinin/Substance P has been implicated in aggression in mammals, including humans. Thus, the higher aggressiveness of Drosophila males reflects the sexually dimorphic expression of a neuropeptide that controls agonistic behaviors across phylogeny.
Collapse
Affiliation(s)
- Kenta Asahina
- Howard Hughes Medical Institute, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA; Division of Biology, 156-29, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| | - Kiichi Watanabe
- Howard Hughes Medical Institute, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA; Division of Biology, 156-29, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| | - Brian J Duistermars
- Howard Hughes Medical Institute, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA; Division of Biology, 156-29, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| | - Eric Hoopfer
- Howard Hughes Medical Institute, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA; Division of Biology, 156-29, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA; Janelia Farm Research Campus, HHMI, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Carlos Roberto González
- Division of Engineering and Applied Science, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| | - Eyrún Arna Eyjólfsdóttir
- Division of Engineering and Applied Science, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| | - Pietro Perona
- Division of Engineering and Applied Science, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| | - David J Anderson
- Howard Hughes Medical Institute, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA; Division of Biology, 156-29, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA.
| |
Collapse
|
27
|
Pascarella G, Lazarevic D, Plessy C, Bertin N, Akalin A, Vlachouli C, Simone R, Faulkner GJ, Zucchelli S, Kawai J, Daub CO, Hayashizaki Y, Lenhard B, Carninci P, Gustincich S. NanoCAGE analysis of the mouse olfactory epithelium identifies the expression of vomeronasal receptors and of proximal LINE elements. Front Cell Neurosci 2014; 8:41. [PMID: 24600346 PMCID: PMC3927265 DOI: 10.3389/fncel.2014.00041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/28/2014] [Indexed: 11/13/2022] Open
Abstract
By coupling laser capture microdissection to nanoCAGE technology and next-generation sequencing we have identified the genome-wide collection of active promoters in the mouse Main Olfactory Epithelium (MOE). Transcription start sites (TSSs) for the large majority of Olfactory Receptors (ORs) have been previously mapped increasing our understanding of their promoter architecture. Here we show that in our nanoCAGE libraries of the mouse MOE we detect a large number of tags mapped in loci hosting Type-1 and Type-2 Vomeronasal Receptors genes (V1Rs and V2Rs). These loci also show a massive expression of Long Interspersed Nuclear Elements (LINEs). We have validated the expression of selected receptors detected by nanoCAGE with in situ hybridization, RT-PCR and qRT-PCR. This work extends the repertory of receptors capable of sensing chemical signals in the MOE, suggesting intriguing interplays between MOE and VNO for pheromone processing and positioning transcribed LINEs as candidate regulatory RNAs for VRs expression.
Collapse
Affiliation(s)
- Giovanni Pascarella
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy ; RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Dejan Lazarevic
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy ; Cluster in Biomedicine (CBM), AREA Science Park Trieste, Italy
| | - Charles Plessy
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Nicolas Bertin
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Altuna Akalin
- Bergen Center for Computational Science - Computational Biology Unit and Sars Centre for Marine Molecular Biology, University of Bergen Bergen, Norway
| | - Christina Vlachouli
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy
| | - Roberto Simone
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy
| | - Geoffrey J Faulkner
- Cancer Biology Program, Mater Medical Research Institute South Brisbane, QLD, Australia ; School of Biomedical Sciences, University of Queensland Brisbane, QLD, Australia
| | - Silvia Zucchelli
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy ; Department of Health Sciences, University of Eastern Piedmont "A. Avogadro," Novara, Italy
| | - Jun Kawai
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Carsten O Daub
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Yoshihide Hayashizaki
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Boris Lenhard
- Bergen Center for Computational Science - Computational Biology Unit and Sars Centre for Marine Molecular Biology, University of Bergen Bergen, Norway
| | - Piero Carninci
- RIKEN Yokohama Institute, Center for Life Science Technologies, Division of Genomic Technologies Tsurumi-ku, Yokohama, Japan
| | - Stefano Gustincich
- Area of Neuroscience, International School for Advanced Studies (SISSA) Trieste, Italy
| |
Collapse
|
28
|
Overath P, Sturm T, Rammensee HG. Of volatiles and peptides: in search for MHC-dependent olfactory signals in social communication. Cell Mol Life Sci 2014; 71:2429-42. [PMID: 24496643 PMCID: PMC4055862 DOI: 10.1007/s00018-014-1559-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/06/2014] [Indexed: 01/11/2023]
Abstract
Genes of the major histocompatibility complex (MHC), which play a critical role in immune recognition, are considered to influence social behaviors in mice, fish, humans, and other vertebrates via olfactory cues. As studied most extensively in mice, the polymorphism of MHC class I genes is considered to bring about a specific scent signature, which is decoded by the olfactory system resulting in an individual-specific reaction such as mating. On the assumption that this signature resides in volatiles, extensive attempts to identify these MHC-specific components in urine failed. Alternatively, it has been suggested that peptide ligands of MHC class I molecules are released into urine and can elicit an MHC-haplotype-specific behavioral response after uptake into the nose by sniffing. Analysis of the urinary peptide composition of mice shows that MHC-derived peptides are present, albeit in extremely low concentrations. In contrast, urine contains abundant peptides which differ between mouse strains due to genomic variations such as single-nucleotide variations or complex polymorphisms in multigene families as well as in their concentration. Thus, urinary peptides represent a real-time sampling of the expressed genome available for sensory evaluation. It is suggested that peptide variation caused by genomic differences contains sufficient information for individual recognition beyond or instead of an influence of the MHC in mice and other vertebrates.
Collapse
Affiliation(s)
- Peter Overath
- Interfakultäres Institut für Zellbiologie, Abteilung Immunologie, Universität Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany,
| | | | | |
Collapse
|
29
|
Ibarra-Soria X, Levitin MO, Logan DW. The genomic basis of vomeronasal-mediated behaviour. Mamm Genome 2013; 25:75-86. [PMID: 23884334 PMCID: PMC3916702 DOI: 10.1007/s00335-013-9463-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 06/19/2013] [Indexed: 11/04/2022]
Abstract
The vomeronasal organ (VNO) is a chemosensory subsystem found in the nose of most mammals. It is principally tasked with detecting pheromones and other chemical signals that initiate innate behavioural responses. The VNO expresses subfamilies of vomeronasal receptors (VRs) in a cell-specific manner: each sensory neuron expresses just one or two receptors and silences all the other receptor genes. VR genes vary greatly in number within mammalian genomes, from no functional genes in some primates to many hundreds in rodents. They bind semiochemicals, some of which are also encoded in gene families that are coexpanded in species with correspondingly large VR repertoires. Protein and peptide cues that activate the VNO tend to be expressed in exocrine tissues in sexually dimorphic, and sometimes individually variable, patterns. Few chemical ligand–VR–behaviour relationships have been fully elucidated to date, largely due to technical difficulties in working with large, homologous gene families with high sequence identity. However, analysis of mouse lines with mutations in genes involved in ligand–VR signal transduction has revealed that the VNO mediates a range of social behaviours, including male–male and maternal aggression, sexual attraction, lordosis, and selective pregnancy termination, as well as interspecific responses such as avoidance and defensive behaviours. The unusual logic of VR expression now offers an opportunity to map the specific neural circuits that drive these behaviours.
Collapse
Affiliation(s)
- Ximena Ibarra-Soria
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | | | | |
Collapse
|
30
|
Prince JEA, Brignall AC, Cutforth T, Shen K, Cloutier JF. Kirrel3 is required for the coalescence of vomeronasal sensory neuron axons into glomeruli and for male-male aggression. Development 2013; 140:2398-408. [PMID: 23637329 DOI: 10.1242/dev.087262] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The accessory olfactory system controls social and sexual interactions in mice that are crucial for survival. Vomeronasal sensory neurons (VSNs) form synapses with dendrites of second order neurons in glomeruli of the accessory olfactory bulb (AOB). Axons of VSNs expressing the same vomeronasal receptor coalesce into multiple glomeruli within spatially conserved regions of the AOB. Here we examine the role of the Kirrel family of transmembrane proteins in the coalescence of VSN axons within the AOB. We find that Kirrel2 and Kirrel3 are differentially expressed in subpopulations of VSNs and that their expression is regulated by activity. Although Kirrel3 expression is not required for early axonal guidance events, such as fasciculation of the vomeronasal tract and segregation of apical and basal VSN axons in the AOB, it is necessary for proper coalescence of axons into glomeruli. Ablation of Kirrel3 expression results in disorganization of the glomerular layer of the posterior AOB and formation of fewer, larger glomeruli. Furthermore, Kirrel3(-/-) mice display a loss of male-male aggression in a resident-intruder assay. Taken together, our results indicate that differential expression of Kirrels on vomeronasal axons generates a molecular code that dictates their proper coalescence into glomeruli within the AOB.
Collapse
Affiliation(s)
- Janet E A Prince
- Montreal Neurological Institute, Centre for Neuronal Survival, 3801 University, Montréal, Québec, H3A 2B4, Canada
| | | | | | | | | |
Collapse
|
31
|
Olfaction and olfactory-mediated behaviour in psychiatric disease models. Cell Tissue Res 2013; 354:69-80. [DOI: 10.1007/s00441-013-1617-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/12/2013] [Indexed: 12/26/2022]
|
32
|
Sano K, Tsuda MC, Musatov S, Sakamoto T, Ogawa S. Differential effects of site-specific knockdown of estrogen receptor α in the medial amygdala, medial pre-optic area, and ventromedial nucleus of the hypothalamus on sexual and aggressive behavior of male mice. Eur J Neurosci 2013; 37:1308-19. [PMID: 23347260 DOI: 10.1111/ejn.12131] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 12/11/2012] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
Abstract
Testosterone is known to play an important role in the regulation of male-type sexual and aggressive behavior. As an aromatised metabolite of testosterone, estradiol-induced activation of estrogen receptor α (ERα) may be crucial for the induction of these behaviors in male mice. However, the importance of ERα expressed in different nuclei for this facilitatory action of testosterone has not been determined. To investigate this issue, we generated an adeno-associated virus vector expressing a small hairpin RNA targeting ERα to site-specifically knockdown ERα expression. We stereotaxically injected either a control or ERα targeting vector into the medial amygdala, medial pre-optic area (MPOA), or ventromedial nucleus of the hypothalamus (VMN) in gonadally intact male mice. Two weeks after injection, all mice were tested biweekly for sexual and aggressive behavior, alternating between behavior tests each week. We found that suppressing ERα in the MPOA reduced sexual but not aggressive behavior, whereas in the VMN it reduced both behaviors. Knockdown of ERα in the medial amygdala did not alter either behavior. Additionally, it was found that ERα knockdown in the MPOA caused a parallel reduction in the number of neuronal nitric oxide synthase-expressing cells. Taken together, these results indicate that the testosterone facilitatory action on male sexual behavior requires the expression of ERα in both the MPOA and VMN, whereas the testosterone facilitatory action on aggression requires the expression of ERα in only the VMN.
Collapse
Affiliation(s)
- Kazuhiro Sano
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | | | | | | |
Collapse
|
33
|
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
|
34
|
Păunescu TG, Rodriguez S, Benz E, McKee M, Tyszkowski R, Albers MW, Brown D. Loss of the V-ATPase B1 subunit isoform expressed in non-neuronal cells of the mouse olfactory epithelium impairs olfactory function. PLoS One 2012; 7:e45395. [PMID: 23028982 PMCID: PMC3447883 DOI: 10.1371/journal.pone.0045395] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/22/2012] [Indexed: 02/07/2023] Open
Abstract
The vacuolar proton-pumping ATPase (V-ATPase) is the main mediator of intracellular organelle acidification and also regulates transmembrane proton (H(+)) secretion, which is necessary for an array of physiological functions fulfilled by organs such as the kidney, male reproductive tract, lung, bone, and ear. In this study we characterize expression of the V-ATPase in the main olfactory epithelium of the mouse, as well as a functional role for the V-ATPase in odor detection. We report that the V-ATPase localizes to the apical membrane microvilli of olfactory sustentacular cells and to the basolateral membrane of microvillar cells. Plasma membrane V-ATPases containing the B1 subunit isoform are not detected in olfactory sensory neurons or in the olfactory bulb. This precise localization of expression affords the opportunity to ascertain the functional relevance of V-ATPase expression upon innate, odor-evoked behaviors in B1-deficient mice. This animal model exhibits diminished innate avoidance behavior (revealed as a decrease in freezing time and an increase in the number of sniffs in the presence of trimethyl-thiazoline) and diminished innate appetitive behavior (a decrease in time spent investigating the urine of the opposite sex). We conclude that V-ATPase-mediated H(+) secretion in the olfactory epithelium is required for optimal olfactory function.
Collapse
Affiliation(s)
- Teodor G Păunescu
- MGH Center for Systems Biology, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | |
Collapse
|
35
|
Neurons expressing trace amine-associated receptors project to discrete glomeruli and constitute an olfactory subsystem. Proc Natl Acad Sci U S A 2012; 109:13410-5. [PMID: 22837392 DOI: 10.1073/pnas.1206724109] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Some chemoreceptors of the trace amine-associated receptor (TAAR) family detect innately aversive odors and are proposed to activate hardwired olfactory circuits. However, the wiring of TAAR neurons, the regulatory mechanisms of Taar gene choice, and the subcellular localization of TAAR proteins remain unknown. Here, we reveal similarities between neurons expressing TAARs and odorant receptors (ORs), but also unexpected differences. Like ORs, TAARs seem to be monoallelically expressed and localized both in cilia, the site of odor detection, and in axons, where they may participate in guidance. TAAR neurons project to discrete glomeruli predominantly localized to a confined bulb region. Taar expression involves different regulatory logic than OR expression, as neurons choosing a Taar5 knockout allele frequently express a second Taar without silencing the deleted allele. Moreover, the epigenetic signature of OR gene choice is absent from Taar genes. The unique molecular and anatomical features of the TAAR neurons suggest that they constitute a distinct olfactory subsystem.
Collapse
|
36
|
Kohl JV. Human pheromones and food odors: epigenetic influences on the socioaffective nature of evolved behaviors. SOCIOAFFECTIVE NEUROSCIENCE & PSYCHOLOGY 2012; 2:17338. [PMID: 24693349 PMCID: PMC3960071 DOI: 10.3402/snp.v2i0.17338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
BACKGROUND Olfactory cues directly link the environment to gene expression. Two types of olfactory cues, food odors and social odors, alter genetically predisposed hormone-mediated activity in the mammalian brain. METHODS The honeybee is a model organism for understanding the epigenetic link from food odors and social odors to neural networks of the mammalian brain, which ultimately determine human behavior. RESULTS Pertinent aspects that extend the honeybee model to human behavior include bottom-up followed by top-down gene, cell, tissue, organ, organ-system, and organism reciprocity; neurophysiological effects of food odors and of sexually dimorphic, species-specific social odors; a model of motor function required for social selection that precedes sexual selection; and hormonal effects that link current neuroscience to social science affects on the development of animal behavior. CONCLUSION As the psychological influence of food odors and social orders is examined in detail, the socioaffective nature of olfactory cues on the biologically based development of sexual preferences across all species that sexually reproduce becomes clearer.
Collapse
Affiliation(s)
- James V. Kohl
- James V. Kohl, Independent Researcher, 175 Crimson Lane, Epworth, GA 30541, USA. Tel: (706) 455-7967.
| |
Collapse
|
37
|
Imai T. Positional information in neural map development: lessons from the olfactory system. Dev Growth Differ 2012; 54:358-65. [PMID: 22404568 DOI: 10.1111/j.1440-169x.2012.01334.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Positional information is fundamental in development. Although molecular gradients are thought to represent positional information in various systems, the molecular logic used to interpret these gradients remains controversial. In the nervous system, sensory maps are formed in the brain based on gradients of axon guidance molecules. However, it remains unclear how axons find their targets based on relative, not absolute, expression levels of axon guidance receptors. No model solely based on axon-target interactions explains this point. Recent studies in the olfactory system suggested that the neural map formation requires axon-axon interactions, which is known as axon sorting. This review discusses how axon-axon and axon-target interactions interpret molecular gradients and determine the axonal projection sites in neural map formation.
Collapse
Affiliation(s)
- Takeshi Imai
- Laboratory for Sensory Circuit Formation, RIKEN Center for Developmental Biology, Kobe 650-0047 PRESTO, Japan Science and Technology Agency, Saitama 332-0012, Japan.
| |
Collapse
|
38
|
Davison IG, Ehlers MD. Neural circuit mechanisms for pattern detection and feature combination in olfactory cortex. Neuron 2011; 70:82-94. [PMID: 21482358 DOI: 10.1016/j.neuron.2011.02.047] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2011] [Indexed: 11/24/2022]
Abstract
Odors are initially encoded in the brain as a set of distinct physicochemical characteristics but are ultimately perceived as a unified sensory object--a "smell." It remains unclear how chemical features encoded by diverse odorant receptors and segregated glomeruli in the main olfactory bulb (MOB) are assembled into integrated cortical representations. Combining patterned optical microstimulation of MOB with in vivo electrophysiological recordings in anterior piriform cortex (PCx), we assessed how cortical neurons decode complex activity patterns distributed across MOB glomeruli. PCx firing was insensitive to single-glomerulus photostimulation. Instead, individual cells reported higher-order combinations of coactive glomeruli resembling odor-evoked sensory maps. Intracellular recordings revealed a corresponding circuit architecture providing each cortical neuron with weak synaptic input from a distinct subpopulation of MOB glomeruli. PCx neurons thus detect specific glomerular ensembles, providing an explicit neural representation of chemical feature combinations that are the hallmark of complex odor stimuli.
Collapse
Affiliation(s)
- Ian G Davison
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
39
|
Trotier D. Vomeronasal organ and human pheromones. Eur Ann Otorhinolaryngol Head Neck Dis 2011; 128:184-90. [PMID: 21377439 DOI: 10.1016/j.anorl.2010.11.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 11/19/2010] [Indexed: 02/03/2023]
Abstract
For many organisms, pheromonal communication is of particular importance in managing various aspects of reproduction. In tetrapods, the vomeronasal (Jacobson's) organ specializes in detecting pheromones in biological substrates of congeners. This information triggers behavioral changes associated, in the case of certain pheromones, with neuroendocrine correlates. In human embryos, the organ develops and the nerve fibers constitute a substrate for the migration of GnRH-secreting cells from the olfactory placode toward the hypothalamus. After this essential step for subsequent secretion of sex hormones by the anterior hypophysis, the organ regresses and the neural connections disappear. The vomeronasal cavities can still be observed by endoscopy in some adults, but they lack sensory neurons and nerve fibers. The genes which code for vomeronasal receptor proteins and the specific ionic channels involved in the transduction process are mutated and nonfunctional in humans. In addition, no accessory olfactory bulbs, which receive information from the vomeronasal receptor cells, are found. The vomeronasal sensory function is thus nonoperational in humans. Nevertheless, several steroids are considered to be putative human pheromones; some activate the anterior hypothalamus, but the effects observed are not comparable to those in other mammals. The signaling process (by neuronal detection and transmission to the brain or by systemic effect) remains to be clearly elucidated.
Collapse
Affiliation(s)
- D Trotier
- CNRS, INRA, FRE 3295, Neurobiologie Sensorielle, domaine de Vilvert, bâtiment 325, 78350 Jouy-en-Josas, France.
| |
Collapse
|
40
|
Lazarini F, Lledo PM. Is adult neurogenesis essential for olfaction? Trends Neurosci 2011; 34:20-30. [DOI: 10.1016/j.tins.2010.09.006] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 09/14/2010] [Accepted: 09/23/2010] [Indexed: 12/31/2022]
|