Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Curr Biol ; 34(7): 1377-1389.e7, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38423017

ABSTRACT

Escaping from danger is one of the most fundamental survival behaviors for animals. Most freshwater fishes display olfactory alarm reactions in which an injured fish releases putative alarm substances from the skin to notify its shoaling company about the presence of danger. Here, we identified two small compounds in zebrafish skin extract, designated as ostariopterin and daniol sulfate. Ostariopterin is a pterin derivative commonly produced in many freshwater fishes belonging to the Ostariophysi superorder. Daniol sulfate is a novel sulfated bile alcohol specifically present in the Danio species, including zebrafish. Ostariopterin and daniol sulfate activate distinct glomeruli in the olfactory bulb. Zebrafish display robust alarm reactions, composed of darting, freezing, and bottom dwelling, only when they are concomitantly stimulated with ostariopterin and daniol sulfate. These results demonstrate that the fish alarm reaction is driven through a coincidence detection mechanism of the two compounds along the olfactory neural circuitry.


Subject(s)
Cyprinidae , Perciformes , Animals , Zebrafish/physiology , Smell , Olfactory Bulb , Sulfates
2.
Cell Rep ; 42(5): 112398, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37083330

ABSTRACT

Spatiotemporal control of gene expression is important for neural development and function. Here, we show that heterogeneous nuclear ribonucleoprotein (hnRNP) A/B is highly expressed in developing olfactory sensory neurons (OSNs), and its knockout results in reduction in mature OSNs and aberrant targeting of OSN axons to the olfactory bulb. RNA immunoprecipitation analysis reveals that hnRNP A/B binds to a group of mRNAs that are highly related to axon projections and synapse assembly. Approximately 11% of the identified hnRNP A/B targets, including Pcdha and Ncam2, encode cell adhesion molecules. In Hnrnpab knockout mice, PCDHA and NCAM2 levels are significantly reduced at the axon terminals of OSNs. Furthermore, deletion of the hnRNP A/B-recognition motif in the 3' UTR of Pcdha leads to impaired PCDHA expression at the OSN axon terminals. Therefore, we propose that hnRNP A/B facilitates OSN maturation and axon projection by regulating the local expression of its target genes at axon terminals.


Subject(s)
Olfactory Receptor Neurons , Animals , Mice , Axons/metabolism , Mice, Knockout , Neural Cell Adhesion Molecules/metabolism , Neurogenesis/genetics , Olfactory Bulb , Olfactory Receptor Neurons/metabolism , Presynaptic Terminals/metabolism
3.
Sci Rep ; 13(1): 1109, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36670138

ABSTRACT

Accumulating evidence suggests that glutathione loss is closely associated with the progression of neurodegenerative disorders. Here, we found that the neuronal conditional-knockout (KO) of glutamyl-cysteine-ligase catalytic-subunit (GCLC), a rate-limiting enzyme for glutathione synthesis, induced brain atrophy accompanied by neuronal loss and neuroinflammation. GCLC-KO mice showed activation of C1q, which triggers engulfment of neurons by microglia, and disease-associated-microglia (DAM), suggesting that activation of microglia is linked to the neuronal loss. Furthermore, gasdermins, which regulate inflammatory form of cell death, were upregulated in the brains of GCLC-KO mice, suggesting the contribution of pyroptosis to neuronal cell death in these animals. In particular, GSDME-deficiency significantly attenuated the hippocampal atrophy and changed levels of DAM markers in GCLC-KO mice. Finally, we found that the expression of GCLC was decreased around amyloid plaques in AppNL-G-F AD model mice. AppNL-G-F mouse also exhibited inflammatory events similar to GCLC-KO mouse. We propose a mechanism by which a vicious cycle of oxidative stress and neuroinflammation enhances neurodegenerative processes. Furthermore, GCLC-KO mouse will serve as a useful tool to investigate the molecular mechanisms underlying neurodegeneration and in the development of new treatment strategies to address neurodegenerative diseases.


Subject(s)
Gasdermins , Neuroinflammatory Diseases , Mice , Animals , Glutathione/metabolism , Brain/metabolism , Oxidative Stress
4.
Neuron ; 110(15): 2455-2469.e8, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35654036

ABSTRACT

The pheromonal information received by the vomeronasal system plays a crucial role in regulating social behaviors such as aggression in mice. Despite accumulating knowledge of the brain regions involved in aggression, the specific vomeronasal receptors and the exact neural circuits responsible for pheromone-mediated aggression remain unknown. Here, we identified one murine vomeronasal receptor, Vmn2r53, that is activated by urine from males of various strains and is responsible for evoking intermale aggression. We prepared a purified pheromonal fraction and Vmn2r53 knockout mice and applied genetic tools for neuronal activity recording, manipulation, and circuit tracing to decipher the neural mechanisms underlying Vmn2r53-mediated aggression. We found that Vmn2r53-mediated aggression is regulated by specific neuronal populations in the ventral premammillary nucleus and the ventromedial hypothalamic nucleus. Together, our results shed light on the hypothalamic regulation of male aggression mediated by a single vomeronasal receptor.


Subject(s)
Aggression , Vomeronasal Organ , Aggression/physiology , Animals , Hypothalamus , Male , Mice , Neurons/physiology , Pheromones/physiology , Ventromedial Hypothalamic Nucleus , Vomeronasal Organ/physiology
5.
Nat Commun ; 13(1): 556, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115521

ABSTRACT

The vomeronasal system plays an essential role in sensing various environmental chemical cues. Here we show that mice exposed to blood and, consequently, hemoglobin results in the activation of vomeronasal sensory neurons expressing a specific vomeronasal G protein-coupled receptor, Vmn2r88, which is mediated by the interaction site, Gly17, on hemoglobin. The hemoglobin signal reaches the medial amygdala (MeA) in both male and female mice. However, it activates the dorsal part of ventromedial hypothalamus (VMHd) only in lactating female mice. As a result, in lactating mothers, hemoglobin enhances digging and rearing behavior. Manipulation of steroidogenic factor 1 (SF1)-expressing neurons in the VMHd is sufficient to induce the hemoglobin-mediated behaviors. Our results suggest that the oxygen-carrier hemoglobin plays a role as a chemosensory signal, eliciting behavioral responses in mice in a state-dependent fashion.


Subject(s)
Amygdala/metabolism , Biomarkers/blood , Hemoglobins/metabolism , Sensory Receptor Cells/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Vomeronasal Organ/metabolism , Animals , Female , Hemoglobins/genetics , In Situ Hybridization/methods , Lactation , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , beta-Globins/genetics , beta-Globins/metabolism
6.
Brain Nerve ; 73(3): 267-272, 2021 Mar.
Article in Japanese | MEDLINE | ID: mdl-33678618

ABSTRACT

The claustrum is a thin, sheet-like neural structure located beneath the cerebral neocortex, and has reciprocal connections with nearly all neocortical areas. It has been hypothesized to play roles in higher brain functions, such as consciousness, multisensory integration, salience detection, and attentional load allocation. However, its roles in brain physiology have not been precisely elucidated, as only a few experimental studies on claustrum function exist. We established a transgenic mouse line expressing Cre recombinase specifically in a population of claustral excitatory neurons that received inputs from and sent outputs to widespread neocortical areas. The claustral neuronal firing was mostly correlated with the cortical slow-wave activity. In vitro optogenetic stimulation of the claustrum induced excitatory postsynaptic responses in most of the neocortical neurons, however, action potentials were primarily elicited in the inhibitory interneurons. In vivo optogenetic stimulation induced a synchronized Down-state featuring prolonged silencing of neural activity in all layers across widespread cortical areas, followed by Down-to-Up state transition. In contrast, genetic ablation of the claustral neurons led to an attenuation of slow-wave activity in the frontal cortex. These results indicate a crucial role of the claustum in synchronizing inhibitory interneurons across the wide cortical areas for spatiotemporal coordination of slow-wave activity.


Subject(s)
Claustrum , Neocortex , Action Potentials , Animals , Basal Ganglia , Mice , Neurons
7.
Nat Neurosci ; 23(6): 741-753, 2020 06.
Article in English | MEDLINE | ID: mdl-32393895

ABSTRACT

During sleep and awake rest, the neocortex generates large-scale slow-wave (SW) activity. Here, we report that the claustrum coordinates neocortical SW generation. We established a transgenic mouse line that enabled the genetic interrogation of a subpopulation of claustral glutamatergic neurons. These neurons received inputs from and sent outputs to widespread neocortical areas. The claustral neuronal firings mostly correlated with cortical SW activity. In vitro optogenetic stimulation of the claustrum induced excitatory postsynaptic responses in most neocortical neurons, but elicited action potentials primarily in inhibitory interneurons. In vivo optogenetic stimulation induced a synchronized down-state featuring prolonged silencing of neural activity in all layers of many cortical areas, followed by a down-to-up state transition. In contrast, genetic ablation of claustral neurons attenuated SW activity in the frontal cortex. These results demonstrate a crucial role of claustral neurons in synchronizing inhibitory interneurons across wide cortical areas for the spatiotemporal coordination of SW activity.


Subject(s)
Claustrum/physiology , Neocortex/physiology , Sleep, Slow-Wave/physiology , Action Potentials/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , Interneurons/physiology , Mice , Mice, Transgenic , Neural Inhibition/physiology , Neurons/physiology , Optogenetics , T-Box Domain Proteins/genetics
8.
J Vis Exp ; (147)2019 05 02.
Article in English | MEDLINE | ID: mdl-31107457

ABSTRACT

Dendritic filopodia are thin and long protrusions based on the actin filament, and they extend and retract as if searching for a target axon. When the dendritic filopodia establish contact with a target axon, they begin maturing into spines, leading to the formation of a synapse. Telencephalin (TLCN) is abundantly localized in dendritic filopodia and is gradually excluded from spines. Overexpression of TLCN in cultured hippocampal neurons induces dendritic filopodia formation. We showed that telencephalin strongly binds to an extracellular matrix molecule, vitronectin. Vitronectin-coated microbeads induced phagocytic cup formation on neuronal dendrites. In the phagocytic cup, TLCN, TLCN-binding proteins such as phosphorylated Ezrin/Radixin/Moesin (phospho-ERM), and F-actin are accumulated, which suggests that components of the phagocytic cup are similar to those of dendritic filopodia. Thus, we developed a method for purifying the phagocytic cup instead of dendritic filopodia. Magnetic polystyrene beads were coated with vitronectin, which is abundantly present in the culture medium of hippocampal neurons and which induces phagocytic cup formation on neuronal dendrites. After 24 h of incubation, the phagocytic cups were mildly solubilized with detergent and collected using a magnet separator. After washing the beads, the binding proteins were eluted and analyzed by silver staining and Western blotting. In the binding fraction, TLCN and actin were abundantly present. In addition, many proteins identified from the fraction were localized to the dendritic filopodia; thus, we named the binding fraction as the dendritic filopodia-rich fraction. This article describes details regarding the purification method for the dendritic filopodia-rich fraction.


Subject(s)
Cell Fractionation/methods , Dendrites/metabolism , Pseudopodia/metabolism , Actins/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Mice , Synapses/physiology , Vitronectin/metabolism
9.
Nat Commun ; 10(1): 209, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30643144

ABSTRACT

Odorants are recognized by multiple olfactory receptors (ORs) and induce innate behaviors like attraction or aversion via olfactory system in mice. However, a role of an individual OR is unclear. Muscone is recognized by a few ORs including MOR215-1 and MOR214-3, and attracts male mice. Odor preference tests using MOR215-1 knockout mice revealed that MOR215-1 and other OR(s), possibly including MOR214-3, are involved in the attraction. (Z)-5-tetradecen-1-ol (Z5-14:OH) activates ~3 ORs, including Olfr288, and evokes attraction at low levels but aversion at higher levels. Olfr288 knockout mice show no attraction but aversion, suggesting Olfr288 is involved in preference for Z5-14:OH, whereas activation of other low-affinity Z5-14:OH receptors evokes aversion. Each OR appears to send a signal to a neural circuit that possesses distinct valence, leading to a certain behavior. The final output behavior with multiple ORs stimulation is determined by summation (addition or competition) of valences coded by activated ORs.


Subject(s)
Instinct , Mating Preference, Animal/physiology , Olfactory Receptor Neurons/physiology , Receptors, Odorant/physiology , Smell/physiology , Animals , Cycloparaffins/chemistry , Cycloparaffins/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Odorants
10.
Nat Commun ; 9(1): 4463, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367054

ABSTRACT

Mating drive is balanced by a need to safeguard resources for offspring, yet the neural basis for negative regulation of mating remains poorly understood. In rodents, pheromones critically regulate sexual behavior. Here, we observe suppression of adult female sexual behavior in mice by exocrine gland-secreting peptide 22 (ESP22), a lacrimal protein from juvenile mice. ESP22 activates a dedicated vomeronasal receptor, V2Rp4, and V2Rp4 knockout eliminates ESP22 effects on sexual behavior. Genetic tracing of ESP22-responsive neural circuits reveals a critical limbic system connection that inhibits reproductive behavior. Furthermore, V2Rp4 counteracts a highly related vomeronasal receptor, V2Rp5, that detects the male sex pheromone ESP1. Interestingly, V2Rp4 and V2Rp5 are encoded by adjacent genes, yet couple to distinct circuits and mediate opposing effects on female sexual behavior. Collectively, our study reveals molecular and neural mechanisms underlying pheromone-mediated sexual rejection, and more generally, how inputs are routed through olfactory circuits to evoke specific behaviors.


Subject(s)
Limbic System/metabolism , Pheromones/metabolism , Receptors, Pheromone/metabolism , Sexual Behavior, Animal , Vomeronasal Organ/metabolism , Animals , Female , Lacrimal Apparatus/metabolism , Limbic System/cytology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Neurons/metabolism , Pheromones/pharmacology , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Pheromone/deficiency , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology
11.
Article in English | MEDLINE | ID: mdl-30147651

ABSTRACT

Dendritic filopodia are thin, long, and highly mobile protrusions functioning as spine precursors. By contrast with a wealth of knowledge on molecular profiles in spines, little is known about structural and functional proteins present in dendritic filopodia. To reveal the molecular constituents of dendritic filopodia, we developed a new method for biochemical preparation of proteins enriched in dendritic filopodia, by taking advantage of specific and strong binding between a dendritic filopodial membrane protein, telencephalin, and its extracellular matrix ligand, vitronectin. When vitronectin-coated magnetic microbeads were added onto cultured hippocampal neurons, phagocytic cup-like membrane protrusions were formed on dendrites through the binding to telencephalin. Magnetically purified membrane protrusion fraction was subjected to comprehensive mass spectrometric analysis and 319 proteins were identified, many of which were confirmed to be localized to dendritic filopodia. Thus, this study provides a useful resource for studying molecular mechanisms underlying dendritic development, synapse formation, and plasticity.

12.
Curr Biol ; 28(8): 1213-1223.e6, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29606417

ABSTRACT

Rodents use the vomeronasal olfactory system to acquire both inter- and intra-specific information from the external environment and take appropriate actions. For example, urinary proteins from predator species elicit avoidance in mice, while those from male mice attract female mice. In addition to urinary proteins, recent studies have highlighted the importance of lacrimal proteins for intra-specific communications in mice. However, whether the tear fluid of other species also mediates social signals remains unknown. Here, we show that a lacrimal protein in rats (predators of mice), called cystatin-related protein 1 (ratCRP1), activates the vomeronasal system of mice. This protein is specifically produced by adult male rats in a steroid hormone-dependent manner, activates the vomeronasal system of female rats, and enhances stopping behavior. When detected by mice, ratCRP1 activates the medial hypothalamic defensive circuit, resulting in decreased locomotion coupled with lowered body temperature and heart rate. Notably, ratCRP1 is recognized by multiple murine type 2 vomeronasal receptors, including Vmn2r28. CRISPR/Cas9-mediated deletion of vmn2r28 impaired both ratCRP1-induced neural activation of the hypothalamic center and decrease of locomotor activity in mice. Taken together, these data reveal the neural and molecular basis by which a tear fluid compound in rats affects the behavior of mice. Furthermore, our study reveals a case in which a single compound that mediates an intra-specific signal in a predator species also functions as an inter-specific signal in the prey species.


Subject(s)
Eye Proteins/physiology , Vomeronasal Organ/physiology , Amygdala/metabolism , Animals , Cystatins/metabolism , Female , Hypothalamus/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Odorants , Predatory Behavior , Proteins/metabolism , Rats , Rodentia/physiology , Smell/physiology , Species Specificity , Vomeronasal Organ/metabolism
13.
Cell Rep ; 22(5): 1115-1123, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29386100

ABSTRACT

Escape responses to threatening stimuli are vital for survival in all animal species. Larval zebrafish display fast escape responses when exposed to tactile, acoustic, and visual stimuli. However, their behavioral responses to chemosensory stimuli remain unknown. In this study, we found that carbon dioxide (CO2) induced a slow avoidance response, which was distinct from the touch-evoked fast escape response. We identified the gonadotropin-releasing hormone 3-expressing terminal nerve as the CO2 sensor in the nose. Wide-field calcium imaging revealed downstream CO2-activated ensembles of neurons along three distinct neural pathways, olfactory, trigeminal, and habenulo-interpeduncular, further reaching the reticulospinal neurons in the hindbrain. Ablation of the nose, terminal nerve, or trigeminal ganglion resulted in a dramatic decrease in CO2-evoked avoidance responses. These findings demonstrate that the terminal nerve-trigeminal system plays a pivotal role in triggering a slow chemosensory avoidance behavior in the larval zebrafish.


Subject(s)
Avoidance Learning/physiology , Carbon Dioxide/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Neurons/drug effects , Neurons/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Animals , Animals, Genetically Modified , Avoidance Learning/drug effects , Larva , Neural Pathways/drug effects , Neural Pathways/metabolism , Pyrrolidonecarboxylic Acid/metabolism , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism , Zebrafish
14.
Nat Commun ; 8(1): 885, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29026079

ABSTRACT

Individual olfactory sensory neurons express a single odorant receptor gene from either class I genes residing in a single cluster on a single chromosome or class II genes spread over multiple clusters on multiple chromosomes. Here, we identify an enhancer element for mouse class I genes, the J element, that is conserved through mammalian species from the platypus to humans. The J element regulates most class I genes expression by exerting an effect over ~ 3 megabases within the whole cluster. Deletion of the trans J element increases the expression frequencies of class I genes from the intact J allele, indicating that the allelic exclusion of class I genes depends on the activity of the J element. Our data reveal a long-range cis-regulatory element that governs the singular class I gene expression and has been phylogenetically preserved to retain a single cluster organization of class I genes in mammals."Each olfactory sensory neuron expresses a single odorant receptor gene from either class I or class II genes. Here, the authors identify an enhancer for mouse class I genes, that is highly conserved, and regulates most class I genes expression by acting over ~ 3 megabases within the whole cluster."


Subject(s)
Enhancer Elements, Genetic , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/genetics , Animals , Conserved Sequence , Gene Expression Regulation , Mice , Multigene Family , Phylogeny
15.
Neuron ; 95(1): 123-137.e8, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28648498

ABSTRACT

In mice, various instinctive behaviors can be triggered by olfactory input. Despite growing knowledge of the brain regions involved in such behaviors, the organization of the neural circuits that convert olfactory input into stereotyped behavioral output remains poorly understood. Here, we mapped the neural circuit responsible for enhancing sexual receptivity of female mice by a male pheromone, exocrine gland-secreting peptide 1 (ESP1). We revealed specific neural types and pathways by which ESP1 information is conveyed from the peripheral receptive organ to the motor-regulating midbrain via the amygdala-hypothalamus axis. In the medial amygdala, a specific type of projection neurons gated ESP1 signals to the ventromedial hypothalamus (VMH) in a sex-dependent manner. In the dorsal VMH, which has been associated with defensive behaviors, a selective neural subpopulation discriminately mediated ESP1 information from a predator cue. Together, our data illuminate a labeled-line organization for controlling pheromone-mediated sexual behavioral output in female mice.


Subject(s)
Amygdala/metabolism , Hypothalamus/metabolism , Mesencephalon/metabolism , Nerve Net/metabolism , Neurons/metabolism , Proteins/metabolism , Sex Attractants/metabolism , Sexual Behavior, Animal/physiology , Amygdala/cytology , Amygdala/physiology , Animals , Cues , Female , Hypothalamus/cytology , Hypothalamus/physiology , Intercellular Signaling Peptides and Proteins , Male , Mesencephalon/cytology , Mesencephalon/physiology , Mice , Mice, Transgenic , Nerve Net/physiology , Neurons/physiology , Predatory Behavior , Sex Characteristics
16.
Curr Biol ; 27(10): 1437-1447.e4, 2017 May 22.
Article in English | MEDLINE | ID: mdl-28502661

ABSTRACT

Nucleotides released from food sources into environmental water are supposed to act as feeding cues for many fish species. However, it remains unknown how fish can sensitively detect those nucleotides. Here we discover a novel olfactory mechanism for ATP sensing in zebrafish. Upon entering into the nostril, ATP is efficiently converted into adenosine through enzymatic reactions of two ecto-nucleotidases expressed in the olfactory epithelium. Adenosine subsequently activates a small population of olfactory sensory neurons expressing a novel adenosine receptor A2c that is unique to fishes and amphibians. The information is then transmitted to a single glomerulus in the olfactory bulb and further to four regions in higher olfactory centers. These results provide conclusive evidence for a sophisticated enzyme-linked receptor mechanism underlying detection of ATP as a food-derived attractive odorant linking to foraging behavior that is crucial and common to aquatic lower vertebrates.


Subject(s)
Adenosine Triphosphate/metabolism , Olfactory Receptor Neurons/physiology , Smell/physiology , Zebrafish/physiology , Adenosine/metabolism , Adenosine Triphosphatases/metabolism , Animals , Behavior, Animal , Calcium/metabolism , Nose/physiology , Olfactory Mucosa/cytology , Olfactory Mucosa/physiology , Olfactory Receptor Neurons/cytology , Phylogeny , Receptors, Purinergic P1/metabolism
18.
Nat Neurosci ; 19(7): 897-904, 2016 07.
Article in English | MEDLINE | ID: mdl-27239939

ABSTRACT

Pheromones play vital roles for survival and reproduction in various organisms. In many fishes, prostaglandin F2α acts not only as a female reproductive hormone, facilitating ovulation and spawning, but also as a sex pheromone inducing male reproductive behaviors. Here, we unravel the molecular and neural circuit mechanisms underlying the pheromonal action of prostaglandin F2α in zebrafish. Prostaglandin F2α specifically activates two olfactory receptors with different sensitivities and expression in distinct populations of ciliated olfactory sensory neurons. Pheromone information is then transmitted to two ventromedial glomeruli in the olfactory bulb and further to four regions in higher olfactory centers. Mutant male zebrafish deficient in the high-affinity receptor exhibit loss of attractive response to prostaglandin F2α and impairment of courtship behaviors toward female fish. These findings demonstrate the functional significance and activation of selective neural circuitry for the sex pheromone prostaglandin F2α and its cognate olfactory receptor in fish reproductive behavior.


Subject(s)
Olfactory Bulb/metabolism , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/metabolism , Receptors, Prostaglandin/metabolism , Smell/physiology , Animals , Courtship , Dinoprost/metabolism , Olfactory Bulb/drug effects , Pheromones/metabolism , Reproduction/physiology , Sexual Behavior, Animal/physiology , Zebrafish
19.
Endocrinology ; 157(4): 1408-20, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26789235

ABSTRACT

Endocrine-disrupting chemicals are prevalent in the environment and can impair reproductive success by affecting the hypothalamic-pituitary-gonadal axis. The developing pituitary gland is sensitive to exposure to endocrine-disrupting chemicals, such as bisphenol A (BPA), and sex-specific effects can occur. However, effects on the critical window of neonatal pituitary gland development in mice have not been explored. Therefore, this study determined baseline gene expression in male and female pituitaries and consequences of environmental exposure to 17ß-estradiol (E2) and BPA on transcription of genes exhibiting sex differences during the neonatal period. Through microarray and quantitative RT-PCR analysis of pituitaries at postnatal day (PND)1, 3 genes were differentially expressed between males and females: Lhb, Fshb, and intracellular adhesion molecule-5 (Icam5). To see whether E2 and BPA exposure regulates these genes, pituitaries were cultured at PND1 with 10(-8) M E2 or 4.4 × 10(-6) M BPA. E2 decreased expression of Lhb, Fshb, and Icam5 mRNA in females but only significantly decreased expression of Icam5 in males. BPA decreased expression of Icam5 similarly to E2, but it did not affect Lhb or Fshb. Importantly, in vivo exposure to 50-µg/kg · d E2 from PND0 to PND7 decreased expression of Lhb, Fshb, and Icam5 mRNA in both males and females, whereas 50-mg/kg · d BPA exposure during the same time frame decreased expression of Icam5 in females only. Overall, we have uncovered that genes differentially expressed between the sexes can be regulated in part by hormonal and chemical signals in vivo and directly at the pituitary and can be regulated in a sex-specific manner.


Subject(s)
Benzhydryl Compounds/pharmacology , Estradiol/pharmacology , Gene Expression/drug effects , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Phenols/pharmacology , Pituitary Gland/drug effects , Animals , Animals, Newborn , Estrogens/pharmacology , Female , Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression Profiling/methods , Immunohistochemistry , In Situ Hybridization , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Pituitary Gland/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Time Factors
20.
Neuron ; 86(4): 985-999, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25959733

ABSTRACT

The mammalian eye-to-brain pathway includes more than 20 parallel circuits, each consisting of precise long-range connections between specific sets of retinal ganglion cells (RGCs) and target structures in the brain. The mechanisms that drive assembly of these parallel connections and the functional implications of their specificity remain unresolved. Here we show that in the absence of contactin 4 (CNTN4) or one of its binding partners, amyloid precursor protein (APP), a subset of direction-selective RGCs fail to target the nucleus of the optic tract (NOT)--the accessory optic system (AOS) target controlling horizontal image stabilization. Conversely, ectopic expression of CNTN4 biases RGCs to arborize in the NOT, and that process also requires APP. Our data reveal critical and novel roles for CNTN4/APP in promoting target-specific axon arborization, and they highlight the importance of this process for functional development of a behaviorally relevant parallel visual pathway.


Subject(s)
Axons/metabolism , Contactins/metabolism , Retina/metabolism , Visual Pathways/physiology , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Mice, Transgenic , Retinal Ganglion Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...