151
|
Distribution of the neuronal inputs to the ventral premammillary nucleus of male and female rats. Brain Res 2014; 1582:77-90. [PMID: 25084037 DOI: 10.1016/j.brainres.2014.07.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 07/17/2014] [Accepted: 07/22/2014] [Indexed: 02/08/2023]
Abstract
The ventral premammillary nucleus (PMV) expresses dense collections of sex steroid receptors and receptors for metabolic cues, including leptin, insulin and ghrelin. The PMV responds to opposite sex odor stimulation and projects to areas involved in reproductive control, including direct innervation of gonadotropin releasing hormone neurons. Thus, the PMV is well positioned to integrate metabolic and reproductive cues, and control downstream targets that mediate reproductive function. In fact, lesions of PMV neurons blunt female reproductive function and maternal aggression. However, although the projections of PMV neurons have been well documented, little is known about the neuronal inputs received by PMV neurons. To fill this gap, we performed a systematic evaluation of the brain sites innervating the PMV neurons of male and female rats using the retrograde tracer subunit B of the cholera toxin (CTb). In general, we observed that males and females show a similar pattern of afferents. We also noticed that the PMV is preferentially innervated by neurons located in the forebrain, with very few projections coming from brainstem nuclei. The majority of inputs originated from the medial nucleus of the amygdala, the bed nucleus of the stria terminalis and the medial preoptic nucleus. A moderate to high density of afferents was also observed in the ventral subiculum, the arcuate nucleus and the ventrolateral subdivision of the ventromedial nucleus of the hypothalamus. Our findings strengthen the concept that the PMV is part of the vomeronasal system and integrates the brain circuitry controlling reproductive functions.
Collapse
|
152
|
The interplay between reproductive social stimuli and adult olfactory bulb neurogenesis. Neural Plast 2014; 2014:497657. [PMID: 25140258 PMCID: PMC4130132 DOI: 10.1155/2014/497657] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 06/19/2014] [Indexed: 12/11/2022] Open
Abstract
Adult neurogenesis is a striking form of structural plasticity that adapts the brain to the changing world. Accordingly, new neuron production is involved in cognitive functions, such as memory, learning, and pattern separation. Recent data in rodents indicate a close link between adult neurogenesis and reproductive social behavior. This provides a key to unravel the functional meaning of adult neurogenesis in biological relevant contexts and, in parallel, opens new perspectives to explore the way the brain is processing social stimuli. In this paper we will summarize some of the major achievements on cues and mechanisms modulating adult neurogenesis during social behaviors related to reproduction and possible role/s played by olfactory newborn neurons in this context. We will point out that newborn interneurons in the accessory olfactory bulb (AOB) represent a privileged cellular target for social stimuli that elicit reproductive behaviors and that such cues modulate adult neurogenesis at two different levels increasing both proliferation of neuronal progenitors in the germinative regions and integration of newborn neurons into functional circuits. This dual mechanism provides fresh neurons that can be involved in critical activities for the individual fitness, that is, the processing of social stimuli driving the parental behavior and partner recognition.
Collapse
|
153
|
Barrios AW, Núñez G, Sánchez Quinteiro P, Salazar I. Anatomy, histochemistry, and immunohistochemistry of the olfactory subsystems in mice. Front Neuroanat 2014; 8:63. [PMID: 25071468 PMCID: PMC4094888 DOI: 10.3389/fnana.2014.00063] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 06/23/2014] [Indexed: 11/13/2022] Open
Abstract
The four regions of the murine nasal cavity featuring olfactory neurons were studied anatomically and by labeling with lectins and relevant antibodies with a view to establishing criteria for the identification of olfactory subsystems that are readily applicable to other mammals. In the main olfactory epithelium and the septal organ the olfactory sensory neurons (OSNs) are embedded in quasi-stratified columnar epithelium; vomeronasal OSNs are embedded in epithelium lining the medial interior wall of the vomeronasal duct and do not make contact with the mucosa of the main nasal cavity; and in Grüneberg's ganglion a small isolated population of OSNs lies adjacent to, but not within, the epithelium. With the exception of Grüneberg's ganglion, all the tissues expressing olfactory marker protein (OMP) (the above four nasal territories, the vomeronasal and main olfactory nerves, and the main and accessory olfactory bulbs) are also labeled by Lycopersicum esculentum agglutinin, while Ulex europaeus agglutinin I labels all and only tissues expressing Gαi2 (the apical sensory neurons of the vomeronasal organ, their axons, and their glomerular destinations in the anterior accessory olfactory bulb). These staining patterns of UEA-I and LEA may facilitate the characterization of olfactory anatomy in other species. A 710-section atlas of the anatomy of the murine nasal cavity has been made available on line.
Collapse
Affiliation(s)
- Arthur W Barrios
- Unit of Anatomy and Embryology, Department of Anatomy and Animal Production, Faculty of Veterinary, University of Santiago de Compostela Lugo, Spain
| | | | - Pablo Sánchez Quinteiro
- Unit of Anatomy and Embryology, Department of Anatomy and Animal Production, Faculty of Veterinary, University of Santiago de Compostela Lugo, Spain
| | - Ignacio Salazar
- Unit of Anatomy and Embryology, Department of Anatomy and Animal Production, Faculty of Veterinary, University of Santiago de Compostela Lugo, Spain
| |
Collapse
|
154
|
Harris JA, Hirokawa KE, Sorensen SA, Gu H, Mills M, Ng LL, Bohn P, Mortrud M, Ouellette B, Kidney J, Smith KA, Dang C, Sunkin S, Bernard A, Oh SW, Madisen L, Zeng H. Anatomical characterization of Cre driver mice for neural circuit mapping and manipulation. Front Neural Circuits 2014; 8:76. [PMID: 25071457 PMCID: PMC4091307 DOI: 10.3389/fncir.2014.00076] [Citation(s) in RCA: 328] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 06/18/2014] [Indexed: 01/26/2023] Open
Abstract
Significant advances in circuit-level analyses of the brain require tools that allow for labeling, modulation of gene expression, and monitoring and manipulation of cellular activity in specific cell types and/or anatomical regions. Large-scale projects and individual laboratories have produced hundreds of gene-specific promoter-driven Cre mouse lines invaluable for enabling genetic access to subpopulations of cells in the brain. However, the potential utility of each line may not be fully realized without systematic whole brain characterization of transgene expression patterns. We established a high-throughput in situ hybridization (ISH), imaging and data processing pipeline to describe whole brain gene expression patterns in Cre driver mice. Currently, anatomical data from over 100 Cre driver lines are publicly available via the Allen Institute's Transgenic Characterization database, which can be used to assist researchers in choosing the appropriate Cre drivers for functional, molecular, or connectional studies of different regions and/or cell types in the brain.
Collapse
Affiliation(s)
| | | | | | - Hong Gu
- Allen Institute for Brain Science Seattle, WA, USA
| | - Maya Mills
- Allen Institute for Brain Science Seattle, WA, USA
| | - Lydia L Ng
- Allen Institute for Brain Science Seattle, WA, USA
| | - Phillip Bohn
- Allen Institute for Brain Science Seattle, WA, USA
| | | | | | | | | | - Chinh Dang
- Allen Institute for Brain Science Seattle, WA, USA
| | - Susan Sunkin
- Allen Institute for Brain Science Seattle, WA, USA
| | - Amy Bernard
- Allen Institute for Brain Science Seattle, WA, USA
| | | | | | - Hongkui Zeng
- Allen Institute for Brain Science Seattle, WA, USA
| |
Collapse
|
155
|
Allen E, Ren J, Zhang Y, Alcedo J. Sensory systems: their impact on C. elegans survival. Neuroscience 2014; 296:15-25. [PMID: 24997267 DOI: 10.1016/j.neuroscience.2014.06.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 06/21/2014] [Accepted: 06/24/2014] [Indexed: 12/24/2022]
Abstract
An animal's survival strongly depends on a nervous system that can rapidly process and integrate the changing quality of its environment and promote the most appropriate physiological responses. This is amply demonstrated in the nematode worm Caenorhabditis elegans, where its sensory system has been shown to impact multiple physiological traits that range from behavior and developmental plasticity to longevity. Because of the accessibility of its nervous system and the number of tools available to study and manipulate its neural circuitry, C. elegans has thus become an important model organism in dissecting the mechanisms through which the nervous system promotes survival. Here we review our current understanding of how the C. elegans sensory system affects diverse physiological traits, whose coordination would be essential for survival under fluctuating environments. The knowledge we derive from the C. elegans studies should provide testable hypotheses in discovering similar mechanisms in higher animals.
Collapse
Affiliation(s)
- Erika Allen
- Department of Biological Sciences, Wayne State University, Detroit, MI 48334, USA
| | - Jing Ren
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Joy Alcedo
- Department of Biological Sciences, Wayne State University, Detroit, MI 48334, USA
| |
Collapse
|
156
|
Abstract
Our ability to understand the function of the nervous system is dependent upon defining the connections of its constituent neurons. Development of methods to define connections within neural networks has always been a growth industry in the neurosciences. Transneuronal spread of neurotropic viruses currently represents the best means of defining synaptic connections within neural networks. The method exploits the ability of viruses to invade neurons, replicate, and spread through the intimate synaptic connections that enable communication among neurons. Since the method was first introduced in the 1970s, it has benefited from an increased understanding of the virus life cycle, the function of viral genome, and the ability to manipulate the viral genome in support of directional spread of virus and the expression of transgenes. In this unit, we review these advances in viral tracing technology and the way in which they may be applied for functional dissection of neural networks.
Collapse
Affiliation(s)
- J Patrick Card
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
157
|
Primary cilia enhance kisspeptin receptor signaling on gonadotropin-releasing hormone neurons. Proc Natl Acad Sci U S A 2014; 111:10335-40. [PMID: 24982149 DOI: 10.1073/pnas.1403286111] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Most central neurons in the mammalian brain possess an appendage called a primary cilium that projects from the soma into the extracellular space. The importance of these organelles is highlighted by the fact that primary cilia dysfunction is associated with numerous neuropathologies, including hyperphagia-induced obesity, hypogonadism, and learning and memory deficits. Neuronal cilia are enriched for signaling molecules, including certain G protein-coupled receptors (GPCRs), suggesting that neuronal cilia sense and respond to neuromodulators in the extracellular space. However, the impact of cilia on signaling to central neurons has never been demonstrated. Here, we show that the kisspeptin receptor (Kiss1r), a GPCR that is activated by kisspeptin to regulate the onset of puberty and adult reproductive function, is enriched in cilia projecting from mouse gonadotropin-releasing hormone (GnRH) neurons. Interestingly, GnRH neurons in adult animals are multiciliated and the percentage of GnRH neurons possessing multiple Kiss1r-positive cilia increases during postnatal development in a progression that correlates with sexual maturation. Remarkably, disruption of cilia selectively on GnRH neurons leads to a significant reduction in kisspeptin-mediated GnRH neuronal activity. To our knowledge, this result is the first demonstration of cilia disruption affecting central neuronal activity and highlights the importance of cilia for proper GPCR signaling.
Collapse
|
158
|
Abstract
Sexually dimorphic behaviors, qualitative or quantitative differences in behaviors between the sexes, result from the activity of a sexually differentiated nervous system. Sensory cues and sex hormones control the entire repertoire of sexually dimorphic behaviors, including those commonly thought to be charged with emotion such as courtship and aggression. Such overarching control mechanisms regulate distinct genes and neurons that in turn specify the display of these behaviors in a modular manner. How such modular control is transformed into cohesive internal states that correspond to sexually dimorphic behavior is poorly understood. We summarize current understanding of the neural circuit control of sexually dimorphic behaviors from several perspectives, including how neural circuits in general, and sexually dimorphic neurons in particular, can generate sexually dimorphic behaviors, and how molecular mechanisms and evolutionary constraints shape these behaviors. We propose that emergent themes such as the modular genetic and neural control of dimorphic behavior are broadly applicable to the neural control of other behaviors.
Collapse
Affiliation(s)
- Cindy F Yang
- Program in Neuroscience, University of California San Francisco, MC2722, San Francisco, CA 94158, USA; Department of Anatomy, University of California San Francisco, MC2722, San Francisco, CA 94158, USA
| | - Nirao M Shah
- Department of Anatomy, University of California San Francisco, MC2722, San Francisco, CA 94158, USA.
| |
Collapse
|
159
|
Abstract
Animal–animal recognition within, and across species, is essential for predator avoidance and social interactions. Despite its essential role in orchestrating responses to animal cues, basic principles of information processing by the vomeronasal system are still unknown. The medial amygdala (MeA) occupies a central position in the vomeronasal pathway, upstream of hypothalamic centers dedicated to defensive and social responses. We have characterized sensory responses in the mouse MeA and uncovered emergent properties that shed new light onto the transformation of vomeronasal information into sex- and species-specific responses. In particular, we show that the MeA displays a degree of stimulus selectivity and a striking sexually dimorphic sensory representation that are not observed in the upstream relay of the accessory olfactory bulb (AOB). Furthermore, our results demonstrate that the development of sexually dimorphic circuits in the MeA requires steroid signaling near the time of puberty to organize the functional representation of sensory stimuli. DOI:http://dx.doi.org/10.7554/eLife.02743.001 Many animals emit and detect chemicals known as pheromones to communicate with other members of their own species. Animals also rely on chemical signals from other species to warn them, for example, that a predator is nearby. Many of these chemical signals—which are present in sweat, tears, urine, and saliva—are detected by a structure called the vomeronasal organ, which is located at the base of the nasal cavity. When this organ detects a particular chemical signal, it broadcasts this information to a network of brain regions that generates an appropriate behavioral response. Two structures within this network, the accessory olfactory bulb and the medial amygdala, play an important role in modifying this signal before it reaches its final destination—a region of the brain called the hypothalamus. Activation of the hypothalamus by the signal triggers changes in the animal's behavior. Although the anatomical details of this pathway have been widely studied, it is not clear how information is actually transmitted along it. Now, Bergan et al. have provided insights into this process by recording signals in the brains of anesthetized mice exposed to specific stimuli. Whereas neurons in the accessory olfactory bulb responded similarly in male and female mice, those in the medial amygdala showed a preference for female urine in male mice, and a preference for male urine in the case of females. This is the first direct demonstration of differences in sensory processing in the brains of male and female mammals. These differences are thought to result from the actions of sex hormones, particularly estrogen, on brain circuits during development. Consistent with this, neurons in the medial amygdala of male mice with reduced levels of estrogen showed a reduced preference for female urine compared to control males. Similarly, female mice that had been previously exposed to high levels of estrogen as pups showed a reduced preference for male urine compared to controls. In addition to increasing understanding of how chemical signals—including pheromones—influence the responses of rodents to other animals, the work of Bergan et al. has provided clues to the neural mechanisms that underlie sex-specific differences in behaviors. DOI:http://dx.doi.org/10.7554/eLife.02743.002
Collapse
Affiliation(s)
- Joseph F Bergan
- Molecular and Cellular Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| | - Yoram Ben-Shaul
- School of Medicine, Department of Medical Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Catherine Dulac
- Molecular and Cellular Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| |
Collapse
|
160
|
Bergan JF, Ben-Shaul Y, Dulac C. Sex-specific processing of social cues in the medial amygdala. eLife 2014. [PMID: 24894465 DOI: 10.7554/elife.02743.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Animal-animal recognition within, and across species, is essential for predator avoidance and social interactions. Despite its essential role in orchestrating responses to animal cues, basic principles of information processing by the vomeronasal system are still unknown. The medial amygdala (MeA) occupies a central position in the vomeronasal pathway, upstream of hypothalamic centers dedicated to defensive and social responses. We have characterized sensory responses in the mouse MeA and uncovered emergent properties that shed new light onto the transformation of vomeronasal information into sex- and species-specific responses. In particular, we show that the MeA displays a degree of stimulus selectivity and a striking sexually dimorphic sensory representation that are not observed in the upstream relay of the accessory olfactory bulb (AOB). Furthermore, our results demonstrate that the development of sexually dimorphic circuits in the MeA requires steroid signaling near the time of puberty to organize the functional representation of sensory stimuli.DOI: http://dx.doi.org/10.7554/eLife.02743.001.
Collapse
Affiliation(s)
- Joseph F Bergan
- Molecular and Cellular Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| | - Yoram Ben-Shaul
- School of Medicine, Department of Medical Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Catherine Dulac
- Molecular and Cellular Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| |
Collapse
|
161
|
Chemosensory Communication of Gender through Two Human Steroids in a Sexually Dimorphic Manner. Curr Biol 2014; 24:1091-5. [DOI: 10.1016/j.cub.2014.03.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 03/01/2014] [Accepted: 03/11/2014] [Indexed: 11/24/2022]
|
162
|
Kirilov M, Clarkson J, Liu X, Roa J, Campos P, Porteous R, Schütz G, Herbison AE. Dependence of fertility on kisspeptin-Gpr54 signaling at the GnRH neuron. Nat Commun 2014; 4:2492. [PMID: 24051579 DOI: 10.1038/ncomms3492] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/22/2013] [Indexed: 12/11/2022] Open
Abstract
Signaling between kisspeptin and its receptor, G-protein-coupled receptor 54 (Gpr54), is now recognized as being essential for normal fertility. However, the key cellular location of kisspeptin-Gpr54 signaling is unknown. Here we create a mouse with a GnRH neuron-specific deletion of Gpr54 to assess the role of gonadotropin-releasing hormone (GnRH) neurons. Mutant mice are infertile, fail to go through puberty and exhibit markedly reduced gonadal size and follicle-stimulating hormone levels alongside GnRH neurons that are unresponsive to kisspeptin. In an attempt to rescue the infertile phenotype of global Gpr54⁻/⁻ mutants, we use BAC transgenesis to target Gpr54 to the GnRH neurons. This results in mice with normal puberty onset, estrous cyclicity, fecundity and a recovery of kisspeptin's stimulatory action upon GnRH neurons. Using complimentary cell-specific knockout and knockin approaches we demonstrate here that the GnRH neuron is the key site of kisspeptin-Gpr54 signaling for fertility.
Collapse
Affiliation(s)
- Milen Kirilov
- Molecular Biology of the Cell I, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
163
|
Cheong RY, Porteous R, Chambon P, Abrahám I, Herbison AE. Effects of neuron-specific estrogen receptor (ER) α and ERβ deletion on the acute estrogen negative feedback mechanism in adult female mice. Endocrinology 2014; 155:1418-27. [PMID: 24476134 DOI: 10.1210/en.2013-1943] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The negative feedback mechanism through which 17β-estradiol (E2) acts to suppress the activity of the GnRH neurons remains unclear. Using inducible and cell-specific genetic mouse models, we examined the estrogen receptor (ER) isoforms expressed by neurons that mediate acute estrogen negative feedback. Adult female mutant mice in which ERα was deleted from all neurons in the neonatal period failed to exhibit estrous cycles or negative feedback. Adult mutant female mice with neonatal neuronal ERβ deletion exhibited normal estrous cycles, but a failure of E2 to suppress LH secretion was seen in ovariectomized mice. Mutant mice with a GnRH neuron-selective deletion of ERβ exhibited normal cycles and negative feedback, suggesting no critical role for ERβ in GnRH neurons in acute negative feedback. To examine the adult roles of neurons expressing ERα, an inducible tamoxifen-based Cre-LoxP approach was used to ablate ERα from neurons that express calmodulin kinase IIα in adults. This resulted in mice with no estrous cycles, a normal increase in LH after ovariectomy, but an inability of E2 to suppress LH secretion. Finally, acute administration of ERα- and ERβ-selective agonists to adult ovariectomized wild-type mice revealed that activation of ERα suppressed LH secretion, whereas ERβ agonists had no effect. This study highlights the differences in adult reproductive phenotypes that result from neonatal vs adult ablation of ERα in the brain. Together, these experiments expand previous global knockout studies by demonstrating that neurons expressing ERα are essential and probably sufficient for the acute estrogen negative feedback mechanism in female mice.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Centre for Neuroendocrinology (R.Y.C., R.P., I.A., A.E.H.), Department of Physiology, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand; and Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), 67400 Illkirch, France
| | | | | | | | | |
Collapse
|
164
|
Giacobini P, Parkash J, Campagne C, Messina A, Casoni F, Vanacker C, Langlet F, Hobo B, Cagnoni G, Gallet S, Hanchate NK, Mazur D, Taniguchi M, Mazzone M, Verhaagen J, Ciofi P, Bouret SG, Tamagnone L, Prevot V. Brain endothelial cells control fertility through ovarian-steroid-dependent release of semaphorin 3A. PLoS Biol 2014; 12:e1001808. [PMID: 24618750 PMCID: PMC3949669 DOI: 10.1371/journal.pbio.1001808] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/30/2014] [Indexed: 11/25/2022] Open
Abstract
Endothelial-cell–derived Sema3A promotes axonal outgrowth and plasticity and thereby regulates neurohormone release in the adult rodent brain in response to the ovarian cycle. Neuropilin-1 (Nrp1) guides the development of the nervous and vascular systems, but its role in the mature brain remains to be explored. Here we report that the expression of the 65 kDa isoform of Sema3A, the ligand of Nrp1, by adult vascular endothelial cells, is regulated during the ovarian cycle and promotes axonal sprouting in hypothalamic neurons secreting gonadotropin-releasing hormone (GnRH), the neuropeptide controlling reproduction. Both the inhibition of Sema3A/Nrp1 signaling and the conditional deletion of Nrp1 in GnRH neurons counteract Sema3A-induced axonal sprouting. Furthermore, the localized intracerebral infusion of Nrp1- or Sema3A-neutralizing antibodies in vivo disrupts the ovarian cycle. Finally, the selective neutralization of endothelial-cell Sema3A signaling in adult Sema3aloxP/loxP mice by the intravenous injection of the recombinant TAT-Cre protein alters the amplitude of the preovulatory luteinizing hormone surge, likely by perturbing GnRH release into the hypothalamo-hypophyseal portal system. Our results identify a previously unknown function for 65 kDa Sema3A-Nrp1 signaling in the induction of axonal growth, and raise the possibility that endothelial cells actively participate in synaptic plasticity in specific functional domains of the adult central nervous system, thus controlling key physiological functions such as reproduction. In the developing embryo, endothelial cells release chemotropic signals such as Semaphorin 3A (Sema3A) that, upon activation of its receptor Neuropilin-1 (Nrp1), regulate neuronal migration and axon guidance. However, whether endothelial cells in the adult brain retain the ability to secrete molecules that influence neuronal function is unknown. Here we show in the adult brain of rodents that vascular endothelial cells release Sema3A and that the amount released is regulated by the ovulatory cycle. Sema3A, in turn, promotes the outgrowth of axons of hypothalamic neurons that express Neuropilin-1 towards the endothelial wall of portal blood vessels. These neurons release there the neuropeptide that controls reproduction: gonadotropin-releasing hormone (GnRH). Notably, this endothelial-cell-mediated sprouting of GnRH axons regulates neuropeptide release at a key stage of the estrous cycle, the proestrus, when the surge of GnRH triggers ovulation. Thus, by promoting GnRH axonal growth in the adult brain, Sema3A/Neuropilin-1 plays a pivotal role in orchestrating the central control of reproduction. Our results suggest a model in which vascular endothelial cells are dynamic signaling components that relay peripheral information to the brain to control key physiological functions, including species survival.
Collapse
Affiliation(s)
- Paolo Giacobini
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Jyoti Parkash
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Céline Campagne
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Andrea Messina
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Filippo Casoni
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Charlotte Vanacker
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Fanny Langlet
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Barbara Hobo
- Netherlands institute for Neuroscience, Amsterdam, The Netherlands
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gabriella Cagnoni
- Candiolo Cancer Institute - FPO (IRCCS) and University of Torino, Department of Oncology, Candiolo, Italy
| | - Sarah Gallet
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Naresh Kumar Hanchate
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Danièle Mazur
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Masahiko Taniguchi
- Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Massimiliano Mazzone
- Versalius Research Center, VIB, Laboratory of Molecular Oncology and Angiogenesis, Leuven, Belgium
- KU Keuven, Versalius Research Center, Leuven, Belgium
| | - Joost Verhaagen
- Netherlands institute for Neuroscience, Amsterdam, The Netherlands
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Philippe Ciofi
- INSERM, Neurocentre Magendie, U862, Université de Bordeaux, Bordeaux, France
| | - Sébastien G. Bouret
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
- The Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California, United States of America
| | - Luca Tamagnone
- Candiolo Cancer Institute - FPO (IRCCS) and University of Torino, Department of Oncology, Candiolo, Italy
| | - Vincent Prevot
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
- * E-mail:
| |
Collapse
|
165
|
The neuroinvasive profiles of H129 (herpes simplex virus type 1) recombinants with putative anterograde-only transneuronal spread properties. Brain Struct Funct 2014; 220:1395-420. [PMID: 24585022 DOI: 10.1007/s00429-014-0733-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 02/11/2014] [Indexed: 10/25/2022]
Abstract
The use of viruses as transneuronal tracers has become an increasingly powerful technique for defining the synaptic organization of neural networks. Although a number of recombinant alpha herpesviruses are known to spread selectively in the retrograde direction through neural circuits only one strain, the H129 strain of herpes simplex virus type 1, is reported to selectively spread in the anterograde direction. However, it is unclear from the literature whether there is an absolute block or an attenuation of retrograde spread of H129. Here, we demonstrate efficient anterograde spread, and temporally delayed retrograde spread, of H129 and three novel recombinants. In vitro studies revealed no differences in anterograde and retrograde spread of parental H129 and its recombinants through superior cervical ganglion neurons. In vivo injections of rat striatum revealed a clear bias of anterograde spread, although evidence of deficient retrograde transport was also present. Evidence of temporally delayed retrograde transneuronal spread of H129 in the retina was observed following injection of the lateral geniculate nucleus. The data also demonstrated that three novel recombinants efficiently express unique fluorescent reporters and have the capacity to infect the same neurons in dual infection paradigms. From these experiments we conclude that H129 and its recombinants not only efficiently infect neurons through anterograde transneuronal passage, but also are capable of temporally delayed retrograde transneuronal spread. In addition, the capacity to produce dual infection of projection targets following anterograde transneuronal passage provides an important addition to viral transneuronal tracing technology.
Collapse
|
166
|
Fraser EJ, Shah NM. Complex chemosensory control of female reproductive behaviors. PLoS One 2014; 9:e90368. [PMID: 24587340 PMCID: PMC3938725 DOI: 10.1371/journal.pone.0090368] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/31/2014] [Indexed: 01/10/2023] Open
Abstract
Olfaction exerts a profound influence on reproductive physiology and behavior in many animals, including rodents. Odors are recognized by sensory neurons residing in the main olfactory epithelium (MOE) and the vomeronasal organ (VNO) in mice and many other vertebrates. The relative contributions of the MOE and VNO in the display of female behaviors are not well understood. Mice null for Cnga2 or Trpc2 essentially lack odor-evoked activity in the MOE and VNO, respectively. Using females mutant for one or both of Cnga2 and Trpc2, we find that maternal care is differentially regulated by the MOE and VNO: retrieval of wandering pups requires the MOE and is regulated redundantly by the VNO whereas maternal aggression requires both sensory epithelia to be functional. Female sexual receptivity appears to be regulated by both the MOE and VNO. Trpc2 null females have previously been shown to display male-type mounting towards other males. Remarkably, we find that females double mutant for Cnga2 and Trpc2 continue to mount other males, indicating that the disinhibition of male-type sexual displays observed in Trpc2 null females does not require chemosensory input from a functional MOE. Taken together, our findings reveal a previously unappreciated complexity in the chemosensory control of reproductive behaviors in the female mouse.
Collapse
Affiliation(s)
- Eleanor J. Fraser
- Program in Genetics, Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America
| | - Nirao M. Shah
- Program in Genetics, Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
167
|
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
|
168
|
Shin J, Prescott M, Mair J, Campbell RE. Roles for primary cilia in gonadotrophin-releasing hormone neurones in the mouse. J Neuroendocrinol 2014; 26:18-25. [PMID: 24279821 DOI: 10.1111/jne.12122] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/15/2013] [Accepted: 11/21/2013] [Indexed: 01/03/2023]
Abstract
During embryonic development, gonadotrophin-releasing hormone (GnRH) neurones make an extraordinary migration out of the nose and into the brain where, in adulthood, they drive the pituitary regulation of gonadal function and fertility. Primary cilia are antennae-like, immotile organelles that project from the surface of nearly all cells, including GnRH neurones. Links between defects in primary cilia and a variety of human pathologies have been discovered that suggest a role for primary cilia in embryogenesis and reproductive function. The present study aimed to investigate whether GnRH neurone primary cilia are critical for their embryonic migration and the adult control of fertility. To achieve this, we used a Cre-loxP strategy to selectively disrupt primary cilia by deleting Kif3a, an intraflagellar transport protein family member essential for primary cilia assembly and function, specifically in GnRH neurones. Confocal analysis revealed that, in Kif3a(fl/fl) (WT-Kif3a) controls, all GnRH neurones possessed primary cilia, whereas, in GnRH-Cre(+/-) ;Kif3a(fl/fl) (GnRH-Kif3aKO) mice, 60% of GnRH neurones lacked any evidence of primary cilia and the remaining 40% possessed only stunted primary cilia (< 2 μm). Despite abolishing normal primary cilia assembly in GnRH neurones from embryogenesis, adult GnRH neurone distribution and reproductive function was remarkably normal. The total number of GnRH neurones was the same in GnRH-Kif3aKO and WT-Kif3a controls; however, a significant increase (25%) was identified in the number of GnRH neurones sampled through the midpoint of the rostral pre-optic area in GnRH-Kif3aKO mice (P < 0.05). The time to vaginal opening was not different in GnRH-Kif3aKO mice, although they displayed significantly advanced first oestrus (P < 0.05), and oestrous cycle length was increased (P < 0.05). However, females displayed normal basal levels of luteinising hormone, responded normally to oestrogen-induced negative- and positive-feedback, and displayed normal fecundity. Taken together, these data suggest that primary cilia and associated signal transduction pathways play a role in the topographical distribution and specific functions of GnRH neurones; however, they are not essential for fertility.
Collapse
Affiliation(s)
- J Shin
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | | | | | | |
Collapse
|
169
|
Abstract
Transient receptor potential (TRP) ion channels have been detected in neurons that are part of the neural network controlling reproductive physiology and behavior. In this chapter we will primarily take a look at the classical/canonical TRP (TRPC) channels but will also examine some other members of the TRP channel superfamily in reproductive (neuro)endocrinology. The referenced data suggest that different TRP proteins could play functional roles at different levels of the reproductive pathway. Still, our understanding of TRP channel involvement in (neuro)endocrinology is quite limited. Due to their mechanism of activation and complex regulation, these channels are however ideally suited to be part of the transduction machinery of hormone-secreting cells.
Collapse
Affiliation(s)
- Trese Leinders-Zufall
- Department of Physiology, University of Saarland School of Medicine, 66421, Homburg, Germany,
| | | |
Collapse
|
170
|
Rao DB, Little PB, Sills R. Subsite awareness in neuropathology evaluation of National Toxicology Program (NTP) studies: a review of select neuroanatomical structures with their functional significance in rodents. Toxicol Pathol 2013; 42:487-509. [PMID: 24135464 PMCID: PMC3965620 DOI: 10.1177/0192623313501893] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This review article is designed to serve as an introductory guide in neuroanatomy for toxicologic pathologists evaluating general toxicity studies. The article provides an overview of approximately 50 neuroanatomical subsites and their functional significance across 7 transverse sections of the brain. Also reviewed are 3 sections of the spinal cord, cranial and peripheral nerves (trigeminal and sciatic, respectively), and intestinal autonomic ganglia. The review is limited to the evaluation of hematoxylin and eosin-stained tissue sections, as light microscopic evaluation of these sections is an integral part of the first-tier toxicity screening of environmental chemicals, drugs, and other agents. Prominent neuroanatomical sites associated with major neurological disorders are noted. This guide, when used in conjunction with detailed neuroanatomic atlases, may aid in an understanding of the significance of functional neuroanatomy, thereby improving the characterization of neurotoxicity in general toxicity and safety evaluation studies.
Collapse
Affiliation(s)
- Deepa B. Rao
- Integrated Laboratory Systems, Inc., Research Triangle Park, North Carolina
| | - Peter B. Little
- Consultant, Experimental Pathology Laboratories, Inc., Research Triangle Park, North Carolina
| | - Robert Sills
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| |
Collapse
|
171
|
Knobloch HS, Charlet A, Stoop R, Grinevich V. Viral Vectors for Optogenetics of Hypothalamic Neuropeptides. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/978-1-62703-610-8_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
172
|
Loss of NSCL-2 in gonadotropin releasing hormone neurons leads to reduction of pro-opiomelanocortin neurons in specific hypothalamic nuclei and causes visceral obesity. J Neurosci 2013; 33:10459-70. [PMID: 23785158 DOI: 10.1523/jneurosci.5287-12.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Regulation of sexual reproduction and energy homeostasis are closely interconnected, but only few efforts were made to explore the impact of gonadotropic neurons on metabolic processes. We have used Nscl-2 mutant mice suffering from adult onset of obesity and hypogonadotropic hypogonadism to study effects of gonadotropin releasing hormone (GnRH) neurons on neuronal circuits controlling energy balance. Inactivation of Nscl-2 in GnRH neurons but not in pro-opiomelanocortin (POMC) neurons reduced POMC neurons and increased visceral fat mass, suggesting a critical role of GnRH cells in the regulation of POMC neurons. In contrast, absence of POMC processing in the majority of Nscl-2-deficient POMC neurons had no effect on energy homeostasis. Finally, we investigated the cellular basis of the reduction of GnRH neurons in NSCL-2 mutants using a lineage tracing approach. We found that loss of Nscl-2 results in aberrant migration of GnRH neurons in Nscl-2 mutant mice causing a lineage switch of ectopically located GnRH neurons.
Collapse
|
173
|
Abstract
Mammalian pheromones control a myriad of innate social behaviors and acutely regulate hormone levels. Responses to pheromones are highly robust, reproducible, and stereotyped and likely involve developmentally predetermined neural circuits. Here, I review several facets of pheromone transduction in mammals, including (a) chemosensory receptors and signaling components of the main olfactory epithelium and vomeronasal organ involved in pheromone detection; (b) pheromone-activated neural circuits subject to sex-specific and state-dependent modulation; and (c) the striking chemical diversity of mammalian pheromones, which range from small, volatile molecules and sulfated steroids to large families of proteins. Finally, I review (d) molecular mechanisms underlying various behavioral and endocrine responses, including modulation of puberty and estrous; control of reproduction, aggression, suckling, and parental behaviors; individual recognition; and distinguishing of own species from predators, competitors, and prey. Deconstruction of pheromone transduction mechanisms provides a critical foundation for understanding how odor response pathways generate instinctive behaviors.
Collapse
Affiliation(s)
- Stephen D Liberles
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115;
| |
Collapse
|
174
|
Garcia-Gonzalez D, Murcia-Belmonte V, Clemente D, De Castro F. Olfactory system and demyelination. Anat Rec (Hoboken) 2013; 296:1424-34. [PMID: 23904351 DOI: 10.1002/ar.22736] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 09/18/2012] [Accepted: 11/29/2012] [Indexed: 01/04/2023]
Abstract
Within the central nervous system, the olfactory system represents one of the most exciting scenarios since it presents relevant examples of long-life sustained neurogenesis and continuous axonal outgrowth from the olfactory epithelium with the subsequent plasticity phenomena in the olfactory bulb. The olfactory nerve is composed of nonmyelinated axons with interesting ontogenetic interpretations. However, the centripetal projections from the olfactory bulb are myelinated axons which project to more caudal areas along the lateral olfactory tract. In consequence, demyelination has not been considered as a possible cause of the olfactory symptoms in those diseases in which this sense is impaired. One prototypical example of an olfactory disease is Kallmann syndrome, in which different mutations give rise to combined anosmia and hypogonadotropic hypogonadism, together with different satellite symptoms. Anosmin-1 is the extracellular matrix glycoprotein altered in the X-linked form of this disease, which participates in cell adhesion and migration, and axonal outgrowth in the olfactory system and in other regions of the central nervous system. Recently, we have described a new patho-physiological role of this protein in the absence of spontaneous remyelination in multiple sclerosis. In the present review, we hypothesize about how both main and satellite neurological symptoms of Kallmann syndrome may be explained by alterations in the myelination. We revisit the relationship between the olfactory system and myelin highlighting that minor histological changes should not be forgotten as putative causes of olfactory malfunction.
Collapse
Affiliation(s)
- D Garcia-Gonzalez
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain
| | | | | | | |
Collapse
|
175
|
Jouhanneau M, Szymanski L, Martini M, Ella A, Keller M. Kisspeptin: a new neuronal target of primer pheromones in the control of reproductive function in mammals. Gen Comp Endocrinol 2013; 188:3-8. [PMID: 23523710 DOI: 10.1016/j.ygcen.2013.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/08/2013] [Indexed: 11/17/2022]
Abstract
Pheromones are known to trigger either short-term behavioral responses, usually referred to as "releaser effects", or more long-term physiological changes, known as "primer effects", which especially affect reproductive function at the level of the gonadotrope axis. The precise mechanisms through which pheromones interact with the gonadotrope axis in the hypothalamus is not fully known. We propose that the neuropeptide Kisspeptin, could be a specific target of primer pheromones, allowing these pheromones to modulate the gonadotrope axis and GnRH activity. This emerging hypothesis is discussed in the context of puberty acceleration in female mice and the male effect in female ungulates (sheep or goat). These examples have been chosen to illustrate the diversity of the reproductive contexts in mammals and potential mechanisms affected by primer effects at the level of the gonadotrope axis.
Collapse
Affiliation(s)
- Mélanie Jouhanneau
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
| | | | | | | | | |
Collapse
|
176
|
Portillo W, Antonio-Cabrera E, Camacho FJ, Díaz NF, Paredes RG. Behavioral characterization of non-copulating male mice. Horm Behav 2013; 64:70-80. [PMID: 23673371 DOI: 10.1016/j.yhbeh.2013.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 04/30/2013] [Accepted: 05/02/2013] [Indexed: 11/26/2022]
Abstract
Non-copulating (NC) males are those animals that do not mate in spite of repeated testing with sexually receptive females. They have been observed in several species including rats and mice. The present experiment was designed to perform a detailed behavioral characterization of NC male mice. Thus, we evaluated their sexual incentive motivation for a sexually receptive female or a sexually active male, olfactory preference for volatile and non-volatile odors from females or males, and olfactory discrimination between female and male volatile odors and food related odors (milk versus vinegar). We compared the activity of the accessory olfactory system (AOS) in copulating (C) and NC males in response to estrous bedding using the induction of Fos-immunoreactivity (Fos-IR) as a measure of neuronal activation. We also determined if estradiol or dopamine treatment could induce sexual behavior in NC males. Finally, we compared the testis weight and the number of penile spines in C, NC, and gonadectomized males. In the sexual incentive motivation test C males spend significantly more time in the female incentive zone than in the male incentive zone. On the other hand, NC males spend the same amount of time in both incentive zones. In tests of olfactory preference, NC males spent less time investigating estrous odors than C males. As well, NC males discriminate urine from conspecifics but they spend less time smelling these odors than C males. In addition, no increase in Fos expression is observed in NC males when they are exposed to odors from estrous females. Our data also suggest that the deficits observed in NC males are not due to lower circulating levels of gonadal hormones, because estradiol supplementation does not induce sexual behavior in these animals, and their testis weight and the number of penile spines are normal. The results suggest that NC males are not sexually motivated by the receptive females and their odors.
Collapse
Affiliation(s)
- W Portillo
- Instituto de Neurobiología Universidad Nacional Autónoma de México, México.
| | | | | | | | | |
Collapse
|
177
|
Carluccio A, Contri A, Amendola S, De Angelis E, De Amicis I, Mazzatenta A. Male isolation: A behavioral representation of the pheromonal ‘female effect’ in donkey (Equus asinus). Physiol Behav 2013; 118:1-7. [DOI: 10.1016/j.physbeh.2013.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 04/11/2013] [Accepted: 04/18/2013] [Indexed: 11/29/2022]
|
178
|
Manoli DS, Fan P, Fraser EJ, Shah NM. Neural control of sexually dimorphic behaviors. Curr Opin Neurobiol 2013; 23:330-8. [PMID: 23680385 DOI: 10.1016/j.conb.2013.04.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/06/2013] [Indexed: 01/18/2023]
Abstract
All sexually reproducing animals exhibit gender differences in behavior. Such sexual dimorphisms in behavior are most obvious in stereotyped displays that enhance reproductive success such as mating, aggression, and parental care. Sexually dimorphic behaviors are a consequence of a sexually differentiated nervous system, and recent studies in fruit flies and mice reveal novel insights into the neural mechanisms that control these behaviors. In the main, these include a diverse array of novel sex differences in the nervous system, surprisingly modular control of various stereotyped dimorphic behavioral routines, and unanticipated sensory and central modulation of mating. We start with a brief overview to provide the appropriate conceptual framework so that the advances made by the newer studies discussed subsequently can be fully appreciated. We restrict our review to reporting progress in understanding the basis of mating and aggression in fruit flies and mice.
Collapse
Affiliation(s)
- Devanand S Manoli
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|
179
|
Alcedo J, Flatt T, Pasyukova EG. Neuronal inputs and outputs of aging and longevity. Front Genet 2013; 4:71. [PMID: 23653632 PMCID: PMC3644678 DOI: 10.3389/fgene.2013.00071] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/13/2013] [Indexed: 12/14/2022] Open
Abstract
An animal’s survival strongly depends on its ability to maintain homeostasis in response to the changing quality of its external and internal environment. This is achieved through intracellular and intercellular communication within and among different tissues. One of the organ systems that plays a major role in this communication and the maintenance of homeostasis is the nervous system. Here we highlight different aspects of the neuronal inputs and outputs of pathways that affect aging and longevity. Accordingly, we discuss how sensory inputs influence homeostasis and lifespan through the modulation of different types of neuronal signals, which reflects the complexity of the environmental cues that affect physiology. We also describe feedback, compensatory, and feed-forward mechanisms in these longevity-modulating pathways that are necessary for homeostasis. Finally, we consider the temporal requirements for these neuronal processes and the potential role of natural genetic variation in shaping the neurobiology of aging.
Collapse
Affiliation(s)
- Joy Alcedo
- Friedrich Miescher Institute for Biomedical Research Basel, Switzerland ; Department of Biological Sciences, Wayne State University Detroit, MI, USA
| | | | | |
Collapse
|
180
|
Petrulis A. Chemosignals, hormones and mammalian reproduction. Horm Behav 2013; 63:723-41. [PMID: 23545474 PMCID: PMC3667964 DOI: 10.1016/j.yhbeh.2013.03.011] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 03/18/2013] [Accepted: 03/21/2013] [Indexed: 11/21/2022]
Abstract
Many mammalian species use chemosignals to coordinate reproduction by altering the physiology and behavior of both sexes. Chemosignals prime reproductive physiology so that individuals become sexually mature and active at times when mating is most probable and suppress it when it is not. Once in reproductive condition, odors produced and deposited by both males and females are used to find and select individuals for mating. The production, dissemination and appropriate responses to these cues are modulated heavily by organizational and activational effects of gonadal sex steroids and thereby intrinsically link chemical communication to the broader reproductive context. Many compounds have been identified as "pheromones" but very few have met the expectations of that term: a unitary, species-typical substance that is both necessary and sufficient for an experience-independent behavioral or physiological response. In contrast, most responses to chemosignals are dependent or heavily modulated by experience, either in adulthood or during development. Mechanistically, chemosignals are perceived by both main and accessory (vomeronasal) olfactory systems with the importance of each system tied strongly to the nature of the stimulus rather than to the response. In the central nervous system, the vast majority of responses to chemosignals are mediated by cortical and medial amygdala connections with hypothalamic and other forebrain structures. Despite the importance of chemosignals in mammals, many details of chemical communication differ even among closely related species and defy clear categorization. Although generating much research and public interest, strong evidence for the existence of a robust chemical communication among humans is lacking.
Collapse
Affiliation(s)
- Aras Petrulis
- Georgia State University, Neuroscience Institute, Atlanta, GA 30303, USA.
| |
Collapse
|
181
|
Abstract
Neural processes that direct an animal's actions toward environmental goals are critical elements for understanding behavior. The hypothalamus is closely associated with motivated behaviors required for survival and reproduction. Intense feeding, drinking, aggressive, and sexual behaviors can be produced by a simple neuronal stimulus applied to discrete hypothalamic regions. What can these "evoked behaviors" teach us about the neural processes that determine behavioral intent and intensity? Small populations of neurons sufficient to evoke a complex motivated behavior may be used as entry points to identify circuits that energize and direct behavior to specific goals. Here, I review recent applications of molecular genetic, optogenetic, and pharmacogenetic approaches that overcome previous limitations for analyzing anatomically complex hypothalamic circuits and their interactions with the rest of the brain. These new tools have the potential to bridge the gaps between neurobiological and psychological thinking about the mechanisms of complex motivated behavior.
Collapse
Affiliation(s)
- Scott M Sternson
- Janelia Farm Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA.
| |
Collapse
|
182
|
Charpentier MJE, Mboumba S, Ditsoga C, Drea CM. Nasopalatine ducts and flehmen behavior in the mandrill: reevaluating olfactory communication in Old World primates. Am J Primatol 2013; 75:703-14. [PMID: 23526642 DOI: 10.1002/ajp.22146] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 02/04/2013] [Accepted: 02/08/2013] [Indexed: 11/06/2022]
Abstract
Compared to other modes of communication, chemical signaling between conspecifics generally has been overlooked in Old World primates, despite the presence in this group of secretory glands and scent-marking behavior, as well as the confirmed production and perception of olfactory signals. In other mammalian species, flehmen is a behavior thought to transport primarily nonvolatile, aqueous-soluble odorants via specialized ducts to the vomeronasal organ (VNO). By contrast, Old World primates are traditionally thought to lack a functional VNO, relying instead on the main olfactory system to process volatile odorants from their environment. Here, in the mandrill (Mandrillus sphinx), we document unusual morphological and behavioral traits that typically are associated with the uptake of conspecific chemical cues for processing by an accessory olfactory system. Notably, we confirmed that both sexes possess open nasopalatine ducts and, in response to the presentation of conspecific odorants, we found that both sexes showed stereotyped behavior consistent with the flehmen response. If, as in other species, flehmen in the mandrill serves to mediate social or reproductive information, we expected its occurrence to vary with characteristics of either the signaler or receiver. Flehmen, particularly in a given male, occurred most often in response to odorants derived from male, as opposed to female, conspecifics. Moreover, odorants derived during the breeding season elicited more flehmen responses than did odorants collected during the birthing season. Lastly, odorants from reproductively cycling females also elicited more responses than did odorants from contracepted females. Although confirming a link between the nasopalatine ducts, flehmen behavior, and olfactory processing in mandrills would require further study, our observations provide new information to suggest anatomical variability within Old World primates, calling further attention to the underappreciated role of chemical communication in this lineage.
Collapse
|
183
|
Suppression of β1-integrin in gonadotropin-releasing hormone cells disrupts migration and axonal extension resulting in severe reproductive alterations. J Neurosci 2013; 32:16992-7002. [PMID: 23175850 DOI: 10.1523/jneurosci.3057-12.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Reproduction in mammals is dependent on the function of hypothalamic neurons whose axons project to the hypothalamic median eminence (ME) where they release gonadotropin-releasing hormone (GnRH) into a specialized capillary network for delivery to the anterior pituitary. These neurons originate prenatally in the nasal placode and migrate into the forebrain along the olfactory-vomeronasal nerves. The complex developmental events leading to the correct establishment of the GnRH system are tightly regulated by the specific spatiotemporal expression patterns of guidance cues and extracellular matrix molecules, the functions of which, in part, are mediated by their binding to β1-subunit-containing integrins. To determine the biological role of these cell-surface proteins in reproduction, Cre/LoxP technology was used to generate GnRH neuron-specific β1-integrin conditional KO (GnRH-Itgb1(-/-)) mice. Loss of β1-integrin signaling impaired migration of GnRH neurons, their axonal extension to the ME, timing of pubertal onset, and fertility in these mice. These results identify β1-integrin as a gene involved in normal development of the GnRH system and demonstrate a fundamental role for this protein in acquisition of normal reproductive competence in female mice.
Collapse
|
184
|
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
|
185
|
Hu Y, Poopalasundaram S, Graham A, Bouloux PM. GnRH neuronal migration and olfactory bulb neurite outgrowth are dependent on FGF receptor 1 signaling, specifically via the PI3K p110α isoform in chick embryo. Endocrinology 2013; 154:388-99. [PMID: 23150492 DOI: 10.1210/en.2012-1555] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fibroblast growth factor (FGF) signaling is essential for both olfactory bulb (OB) morphogenesis and the specification, migration, and maturation of the GnRH-secreting neurons. Disruption of FGF signaling contributes to Kallmann syndrome characterized by both anosmia and sexual immaturity. However, several unanswered questions remain as to which specific FGF receptor (FGFR)-1 signaling pathways are necessary for OB and GnRH neuronal development. Here, using pharmacological phosphatidylinositol 3-kinase (PI3K) isoform-specific inhibitors, we demonstrate a central role for the PI3K p110α isoform as a downstream effector of FGFR1 signaling for both GnRH neuronal migration and OB development. We show that signaling via the PI3K p110α isoform is required for GnRH neuronal migration in explant cultures of embryonic day (E) 4 chick olfactory placodes. We also show that in ovo administration of LY294002, a global PI3K inhibitor as well as an inhibitor to the PI3K p110α isoform into the olfactory placode of E3 chick embryo impairs GnRH neuronal migration toward the forebrain. In contrast, in ovo PI3K inhibitor treatment produced no obvious defects on primary olfactory sensory neuron axonal targeting and bundle formation. We also demonstrate that anosmin-1 and FGF2 induced neuronal migration of immortalized human embryonic GnRH neuroblast cells (FNC-B4-hTERT) is mediated by modulating FGFR1 signaling via the PI3K p110α isoform, specifically through phosphorylation of the PI3K downstream effectors, Akt and glycogen synthase kinase-3β. Finally, we show that neurite outgrowth and elongation of OB neurons in E10 chick OB explants are also dependent on the PI3K p110α isoform downstream of FGFR1. This study provides mechanistic insight into the etiology of Kallmann syndrome.
Collapse
Affiliation(s)
- Youli Hu
- Centre for Neuroendocrinology, University College London Medical School, Royal Free Campus, London NW3 2PF, United Kingdom.
| | | | | | | |
Collapse
|
186
|
Suárez R, García-González D, de Castro F. Mutual influences between the main olfactory and vomeronasal systems in development and evolution. Front Neuroanat 2012; 6:50. [PMID: 23269914 PMCID: PMC3529325 DOI: 10.3389/fnana.2012.00050] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 11/26/2012] [Indexed: 12/14/2022] Open
Abstract
The sense of smell plays a crucial role in the sensory world of animals. Two chemosensory systems have been traditionally thought to play-independent roles in mammalian olfaction. According to this, the main olfactory system (MOS) specializes in the detection of environmental odorants, while the vomeronasal system (VNS) senses pheromones and semiochemicals produced by individuals of the same or different species. Although both systems differ in their anatomy and function, recent evidence suggests they act synergistically in the perception of scents. These interactions include similar responses to some ligands, overlap of telencephalic connections and mutual influences in the regulation of olfactory-guided behavior. In the present work, we propose the idea that the relationships between systems observed at the organismic level result from a constant interaction during development and reflects a common history of ecological adaptations in evolution. We review the literature to illustrate examples of developmental and evolutionary processes that evidence these interactions and propose that future research integrating both systems may shed new light on the mechanisms of olfaction.
Collapse
Affiliation(s)
- Rodrigo Suárez
- Queensland Brain Institute, The University of Queensland, St Lucia Brisbane, QLD, Australia ; Departamento de Biología, Facultad de Ciencias, Universidad de Chile Santiago, Chile
| | | | | |
Collapse
|
187
|
Gascuel J, Lemoine A, Rigault C, Datiche F, Benani A, Penicaud L, Lopez-Mascaraque L. Hypothalamus-olfactory system crosstalk: orexin a immunostaining in mice. Front Neuroanat 2012; 6:44. [PMID: 23162437 PMCID: PMC3492705 DOI: 10.3389/fnana.2012.00044] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 10/06/2012] [Indexed: 01/06/2023] Open
Abstract
It is well known that olfaction influences food intake, and conversely, that an individual’s nutritional status modulates olfactory sensitivity. However, what is still poorly understood is the neuronal correlate of this relationship, as well as the connections between the olfactory bulb and the hypothalamus. The goal of this report is to analyze the relationship between the olfactory bulb and hypothalamus, focusing on orexin A immunostaining, a hypothalamic neuropeptide that is thought to play a role in states of sleep/wakefulness. Interestingly, orexin A has also been described as a food intake stimulator. Such an effect may be due in part to the stimulation of the olfactory bulbar pathway. In rats, orexin positive cells are concentrated strictly in the lateral hypothalamus, while their projections invade nearly the entire brain including the olfactory system. Therefore, orexin appears to be a good candidate to play a pivotal role in connecting olfactory and hypothalamic pathways. So far, orexin has been described in rats, however, there is still a lack of information concerning its expression in the brains of adult and developing mice. In this context, we revisited the orexin A pattern in adult and developing mice using immunohistological methods and confocal microscopy. Besides minor differences, orexin A immunostaining in mice shares many features with those observed in rats. In the olfactory bulb, even though there are few orexin projections, they reach all the different layers of the olfactory bulb. In contrast to the presence of orexin projections in the main olfactory bulb, almost none have been found in the accessory olfactory bulb. The developmental expression of orexin A supports the hypothesis that orexin expression only appears post-natally.
Collapse
Affiliation(s)
- Jean Gascuel
- Instituto Cajal, CSIC, Avda del Doctor Arce Madrid, Spain ; CNRS UMR 6265, Centre des Sciences du Goût et de l'Alimentation Dijon, France ; Institut National de la Recherche Agronomique UMR 1324, Centre des Sciences du Goût et de l'Alimentation Dijon, France ; Université de Bourgogne UMR CSGA, Centre des Sciences du Goût et de l'Alimentation Dijon, France
| | | | | | | | | | | | | |
Collapse
|
188
|
The role of cAMP response element-binding protein in estrogen negative feedback control of gonadotropin-releasing hormone neurons. J Neurosci 2012; 32:11309-17. [PMID: 22895714 DOI: 10.1523/jneurosci.1333-12.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The mechanisms through which estradiol (E2) regulates gonadotropin-releasing hormone (GnRH) neurons to control fertility are unclear. Previous studies have demonstrated that E2 rapidly phosphorylates cAMP response element-binding protein (CREB) in GnRH neurons in vivo. In the present study, we used GnRH neuron-specific CREB-deleted mutant mice [GnRH-CREB knock-outs (KOs)] with and without global cAMP response element modulator (CREM) deletion (global-CREM KOs) to investigate the role of CREB in estrogen negative feedback on GnRH neurons. Evaluation of GnRH-CREB KO mice with and without global CREM deletion revealed normal puberty onset. Although estrus cycle length in adults was the same in controls and knock-out mice, cycles in mutant mice consisted of significantly longer periods of diestrus and less estrus. In GnRH-CREB KO mice, basal levels of luteinizing hormone (LH) and the postovariectomy increment in LH were normal, but the ability of E2 to rapidly suppress LH was significantly blunted. In contrast, basal and postovariectomy LH levels were abnormal in GnRH-CREB KO/global-CREM KO mice. Fecundity studies showed that GnRH-CREB KO with and without global CREM deletion were normal up to ∼9 months of age, at which time they became prematurely reproductively senescent. Morphological analysis of GnRH neurons revealed a significant reduction (p < 0.01) in GnRH somatic spine density of GnRH-CREB KO mice compared to control females. These observations implicate CREB within the GnRH neuron as an important target for E2's negative feedback actions. They also indicate that the rapid modulation of CREB by E2 is of physiological significance in the CNS.
Collapse
|
189
|
Niimi K, Horie S, Yokosuka M, Kawakami-Mori F, Tanaka K, Fukayama H, Sahara Y. Heterogeneous electrophysiological and morphological properties of neurons in the mouse medial amygdala in vitro. Brain Res 2012; 1480:41-52. [PMID: 22960119 DOI: 10.1016/j.brainres.2012.08.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 08/26/2012] [Accepted: 08/27/2012] [Indexed: 10/27/2022]
Abstract
Neurons in the medial nucleus of the amygdala (MeA) play a key role in the innate maternal, reproductive, defensive, and social behaviors. However, it is unclear how activation of the vomeronasal system leads to the behavioral outputs that are associated with pheromones. Here, we characterized the electrophysiological and morphological properties of MeA neurons using whole-cell recordings in mice slice preparations. Biocytin labeling revealed that MeA neurons possessed bipolar to multipolar cell bodies and dendritic fields covering projection areas from the accessory olfactory bulb. In 70% of recorded MeA neurons, monosynaptic excitatory postsynaptic currents (EPSCs) were evoked from the accessory olfactory bulb afferent in which the α-amino-3-hydroxy-5-methyl-4-isoxazole propionate component was dominant and was rarely followed by the N-methyl-d-aspartic acid component. Norepinephrine increased the frequency of spontaneous inhibitory postsynaptic currents in some neurons, whereas α-methyl-5-hydroxytryptamine increased spontaneous EPSCs in other neurons. Morphologically and physiologically, heterogeneous MeA neurons appear likely to produce multiplex outputs of instinctive behaviors.
Collapse
Affiliation(s)
- Keita Niimi
- Departments of Physiology, Tsurumi University School of Dental Medicine, Yokohama 230-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
190
|
Abstract
Persistent alterations in network activity trigger compensatory changes in excitation and inhibition that restore neuronal firing rate to an optimal range. One example of such synaptic homeostasis is the downregulation of inhibitory transmission by chronic inactivity, in part through the reduction of vesicular transmitter content. The enzyme glutamic acid decarboxylase 67 (GAD67) is critical for GABA synthesis, but its involvement in homeostatic plasticity is unclear. We explored the role of GAD67 in activity-dependent synaptic plasticity using a mouse line (Gad1(-/-)) in which GAD67 expression is disrupted by genomic insertion of the green fluorescent protein (GFP). Homozygous deletion of Gad1 significantly reduced miniature inhibitory postsynaptic current (mIPSC) amplitudes and GABA levels in cultured hippocampal neurons. The fractional block of mIPSC amplitude by a low affinity, competitive GABA(A) receptor antagonist was higher in GAD67-lacking neurons, suggesting that GABA concentration in the synaptic cleft is lower in knockout animals. Chronic suppression of activity by the application of tetrodotoxin (TTX) reduced mIPSC amplitudes and the levels of GAD67 and GABA. Moreover, TTX reduced GFP levels in interneurons, suggesting that GAD67 gene expression is a key regulatory target of activity. These in vitro experiments were corroborated by in vivo studies in which olfactory deprivation reduced mIPSC amplitudes and GFP levels in glomerular neurons in the olfactory bulb. Importantly, TTX-induced downregulation of mIPSC was attenuated in Gad1(-/-) neurons. Altogether, these findings indicate that activity-driven expression of GAD67 critically controls GABA synthesis and, thus, vesicular filling of the transmitter.
Collapse
|
191
|
Schauer C, Leinders-Zufall T. Imaging calcium responses in GFP-tagged neurons of hypothalamic mouse brain slices. J Vis Exp 2012:e4213. [PMID: 22951467 DOI: 10.3791/4213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Despite an enormous increase in our knowledge about the mechanisms underlying the encoding of information in the brain, a central question concerning the precise molecular steps as well as the activity of specific neurons in multi-functional nuclei of brain areas such as the hypothalamus remain. This problem includes identification of the molecular components involved in the regulation of various neurohormone signal transduction cascades. Elevations of intracellular Ca(2+) play an important role in regulating the sensitivity of neurons, both at the level of signal transduction and at synaptic sites. New tools have emerged to help identify neurons in the myriad of brain neurons by expressing green fluorescent protein (GFP) under the control of a particular promoter. To monitor both spatially and temporally stimulus-induced Ca(2+) responses in GFP-tagged neurons, a non-green fluorescent Ca(2+) indicator dye needs to be used. In addition, confocal microscopy is a favorite method of imaging individual neurons in tissue slices due to its ability to visualize neurons in distinct planes of depth within the tissue and to limit out-of-focus fluorescence. The ratiometric Ca(2+) indicator fura-2 has been used in combination with GFP-tagged neurons. However, the dye is excited by ultraviolet (UV) light. The cost of the laser and the limited optical penetration depth of UV light hindered its use in many laboratories. Moreover, GFP fluorescence may interfere with the fura-2 signals. Therefore, we decided to use a red fluorescent Ca(2+) indicator dye. The huge Stokes [corrected] shift of fura-red permits multicolor analysis of the red fluorescence in combination with GFP using a single excitation wavelength. We had previously good results using fura-red in combination with GFP-tagged olfactory neurons. The protocols for olfactory tissue slices seemed to work equally well in hypothalamic neurons. Fura-red based Ca(2+) imaging was also successfully combined with GFP-tagged pancreatic β-cells and GFP-tagged receptors expressed in HEK cells. A little quirk of fura-red is that its fluorescence intensity at 650 nm decreases once the indicator binds calcium. Therefore, the fluorescence of resting neurons with low Ca(2+) concentration has relatively high intensity. It should be noted, that other red Ca(2+)-indicator dyes exist or are currently being developed, that might give better or improved results in different neurons and brain areas.
Collapse
Affiliation(s)
- Christian Schauer
- Department of Physiology, School of Medicine, University of Saarland, Homburg, Germany
| | | |
Collapse
|
192
|
Hanchate NK, Giacobini P, Lhuillier P, Parkash J, Espy C, Fouveaut C, Leroy C, Baron S, Campagne C, Vanacker C, Collier F, Cruaud C, Meyer V, García-Piñero A, Dewailly D, Cortet-Rudelli C, Gersak K, Metz C, Chabrier G, Pugeat M, Young J, Hardelin JP, Prevot V, Dodé C. SEMA3A, a gene involved in axonal pathfinding, is mutated in patients with Kallmann syndrome. PLoS Genet 2012; 8:e1002896. [PMID: 22927827 PMCID: PMC3426548 DOI: 10.1371/journal.pgen.1002896] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 07/02/2012] [Indexed: 12/29/2022] Open
Abstract
Kallmann syndrome (KS) associates congenital hypogonadism due to gonadotropin-releasing hormone (GnRH) deficiency and anosmia. The genetics of KS involves various modes of transmission, including oligogenic inheritance. Here, we report that Nrp1sema/sema mutant mice that lack a functional semaphorin-binding domain in neuropilin-1, an obligatory coreceptor of semaphorin-3A, have a KS–like phenotype. Pathohistological analysis of these mice indeed showed abnormal development of the peripheral olfactory system and defective embryonic migration of the neuroendocrine GnRH cells to the basal forebrain, which results in increased mortality of newborn mice and reduced fertility in adults. We thus screened 386 KS patients for the presence of mutations in SEMA3A (by Sanger sequencing of all 17 coding exons and flanking splice sites) and identified nonsynonymous mutations in 24 patients, specifically, a frameshifting small deletion (D538fsX31) and seven different missense mutations (R66W, N153S, I400V, V435I, T688A, R730Q, R733H). All the mutations were found in heterozygous state. Seven mutations resulted in impaired secretion of semaphorin-3A by transfected COS-7 cells (D538fsX31, R66W, V435I) or reduced signaling activity of the secreted protein in the GN11 cell line derived from embryonic GnRH cells (N153S, I400V, T688A, R733H), which strongly suggests that these mutations have a pathogenic effect. Notably, mutations in other KS genes had already been identified, in heterozygous state, in five of these patients. Our findings indicate that semaphorin-3A signaling insufficiency contributes to the pathogenesis of KS and further substantiate the oligogenic pattern of inheritance in this developmental disorder. Kallmann syndrome is a hereditary developmental disease that affects both the hormonal reproductive axis and the sense of smell. There is a developmental link between the reproductive and olfactory disorders: neuroendocrine cells producing the gonadotropin-releasing hormone that is deficient in the patients normally migrate from the nose to the forebrain along olfactory nerve fibers during embryonic life, and they fail to do so in the patients. Affected individuals usually do not undergo spontaneous puberty. Hormone replacement therapy is the treatment to initiate virilization in males or breast development in females and later to develop fertility in both sexes. This is a genetically heterogeneous disease. Mutations in any of eight causative genes identified so far have been found in approximately 30% of the affected individuals, thus indicating that other genes remain to be discovered. We report on the identification, in 6% of the KS patients, of various loss-of-function mutations in the gene coding for semaphorin-3A, a secreted protein involved in the navigation of olfactory nerve fibers during embryogenesis. The fact that many of these mutations were also detected in clinically unaffected individuals indicates that they must combine with other genetic defects to produce the disease phenotype.
Collapse
Affiliation(s)
- Naresh Kumar Hanchate
- Inserm U837, Développement et Plasticité du Cerveau Postnatal, Centre de Recherche Jean-Pierre Aubert, Lille, France
- Université Lille Nord de France, Lille, France
- UDSL, Ecole de Médecine, Lille, France
| | - Paolo Giacobini
- Inserm U837, Développement et Plasticité du Cerveau Postnatal, Centre de Recherche Jean-Pierre Aubert, Lille, France
- Université Lille Nord de France, Lille, France
- UDSL, Ecole de Médecine, Lille, France
| | - Pierre Lhuillier
- Institut Cochin, Département de Génétique et Développement, Inserm U1016, Université Paris-Descartes, Paris, France
| | - Jyoti Parkash
- Inserm U837, Développement et Plasticité du Cerveau Postnatal, Centre de Recherche Jean-Pierre Aubert, Lille, France
- Université Lille Nord de France, Lille, France
- UDSL, Ecole de Médecine, Lille, France
| | - Cécile Espy
- Laboratoire de Biochimie et Génétique Moléculaire, Hôpital Cochin, APHP, Paris, France
| | - Corinne Fouveaut
- Laboratoire de Biochimie et Génétique Moléculaire, Hôpital Cochin, APHP, Paris, France
| | - Chrystel Leroy
- Laboratoire de Biochimie et Génétique Moléculaire, Hôpital Cochin, APHP, Paris, France
| | - Stéphanie Baron
- Laboratoire de Biochimie et Génétique Moléculaire, Hôpital Cochin, APHP, Paris, France
| | - Céline Campagne
- Inserm U837, Développement et Plasticité du Cerveau Postnatal, Centre de Recherche Jean-Pierre Aubert, Lille, France
- Université Lille Nord de France, Lille, France
- UDSL, Ecole de Médecine, Lille, France
| | - Charlotte Vanacker
- Inserm U837, Développement et Plasticité du Cerveau Postnatal, Centre de Recherche Jean-Pierre Aubert, Lille, France
- Université Lille Nord de France, Lille, France
- UDSL, Ecole de Médecine, Lille, France
| | - Francis Collier
- Université Lille Nord de France, Lille, France
- UDSL, Ecole de Médecine, Lille, France
- CHRU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Roger Salengro, Lille, France
| | - Corinne Cruaud
- Institut de Génomique, Genoscope, CEA, DSV, Evry, France
| | - Vincent Meyer
- Institut de Génomique, Genoscope, CEA, DSV, Evry, France
| | | | - Didier Dewailly
- Inserm U837, Développement et Plasticité du Cerveau Postnatal, Centre de Recherche Jean-Pierre Aubert, Lille, France
- Université Lille Nord de France, Lille, France
- UDSL, Ecole de Médecine, Lille, France
- CHRU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Roger Salengro, Lille, France
| | | | - Ksenija Gersak
- Department of Obstetrics and Gynecology, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Chantal Metz
- Pôle Femme, Mère, et Enfant, CHU Morvan, Brest, France
| | | | - Michel Pugeat
- Service d'Endocrinologie, Hôpital Neurologique et Neurochirurgical, Bron, France
| | - Jacques Young
- Service d'Endocrinologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Jean-Pierre Hardelin
- Inserm U587, Département de Neuroscience, Institut Pasteur, Université Pierre et Marie Curie – Paris 06, Paris, France
- * E-mail: (J-P Hardelin); (V Prevot)
| | - Vincent Prevot
- Inserm U837, Développement et Plasticité du Cerveau Postnatal, Centre de Recherche Jean-Pierre Aubert, Lille, France
- Université Lille Nord de France, Lille, France
- UDSL, Ecole de Médecine, Lille, France
- * E-mail: (J-P Hardelin); (V Prevot)
| | - Catherine Dodé
- Institut Cochin, Département de Génétique et Développement, Inserm U1016, Université Paris-Descartes, Paris, France
- Laboratoire de Biochimie et Génétique Moléculaire, Hôpital Cochin, APHP, Paris, France
| |
Collapse
|
193
|
Watabe-Uchida M, Zhu L, Ogawa SK, Vamanrao A, Uchida N. Whole-brain mapping of direct inputs to midbrain dopamine neurons. Neuron 2012; 74:858-73. [PMID: 22681690 DOI: 10.1016/j.neuron.2012.03.017] [Citation(s) in RCA: 917] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2012] [Indexed: 12/27/2022]
Abstract
Recent studies indicate that dopamine neurons in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) convey distinct signals. To explore this difference, we comprehensively identified each area's monosynaptic inputs using the rabies virus. We show that dopamine neurons in both areas integrate inputs from a more diverse collection of areas than previously thought, including autonomic, motor, and somatosensory areas. SNc and VTA dopamine neurons receive contrasting excitatory inputs: the former from the somatosensory/motor cortex and subthalamic nucleus, which may explain their short-latency responses to salient events; and the latter from the lateral hypothalamus, which may explain their involvement in value coding. We demonstrate that neurons in the striatum that project directly to dopamine neurons form patches in both the dorsal and ventral striatum, whereas those projecting to GABAergic neurons are distributed in the matrix compartment. Neuron-type-specific connectivity lays a foundation for studying how dopamine neurons compute outputs.
Collapse
Affiliation(s)
- Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | | | | | | | | |
Collapse
|
194
|
Elias CF, Purohit D. Leptin signaling and circuits in puberty and fertility. Cell Mol Life Sci 2012; 70:841-62. [PMID: 22851226 PMCID: PMC3568469 DOI: 10.1007/s00018-012-1095-1] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 07/05/2012] [Accepted: 07/09/2012] [Indexed: 12/22/2022]
Abstract
Leptin is an adipocyte-derived hormone involved in a myriad of physiological process, including the control of energy balance and several neuroendocrine axes. Leptin-deficient mice and humans are obese, diabetic, and display a series of neuroendocrine and autonomic abnormalities. These individuals are infertile due to a lack of appropriate pubertal development and inadequate synthesis and secretion of gonadotropins and gonadal steroids. Leptin receptors are expressed in many organs and tissues, including those related to the control of reproductive physiology (e.g., the hypothalamus, pituitary gland, and gonads). In the last decade, it has become clear that leptin receptors located in the brain are major players in most leptin actions, including reproduction. Moreover, the recent development of molecular techniques for brain mapping and the use of genetically modified mouse models have generated crucial new findings for understanding leptin physiology and the metabolic influences on reproductive health. In the present review, we will highlight the new advances in the field, discuss the apparent contradictions, and underline the relevance of this complex physiological system to human health. We will focus our review on the hypothalamic circuitry and potential signaling pathways relevant to leptin’s effects in reproductive control, which have been identified with the use of cutting-edge technologies of molecular mapping and conditional knockouts.
Collapse
Affiliation(s)
- Carol F Elias
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Y6-220B, Dallas, TX, 75390-9077, USA.
| | | |
Collapse
|
195
|
Cheong RY, Kwakowsky A, Barad Z, Porteous R, Herbison AE, Ábrahám IM. Estradiol acts directly and indirectly on multiple signaling pathways to phosphorylate cAMP-response element binding protein in GnRH neurons. Endocrinology 2012; 153:3792-803. [PMID: 22719057 DOI: 10.1210/en.2012-1232] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rapid, nonclassical 17β-estradiol (E2) actions are thought to play an important role in the modulation of neuronal function. The present study addresses the intracellular signaling cascades involved in the rapid E2-induced phosphorylation of cAMP response element binding protein (CREB) in GnRH neurons. Administration of E2 to adult female mice resulted in the activation of ERK1/2 in GnRH neurons within 15 min. In vitro studies using pharmacological antagonists showed that ERK1/2 was essential for E2-induced CREB phosphorylation in GnRH neurons. Upstream to this, protein kinase A and calcium/calmodulin-dependent protein kinase type II, but not protein kinase C, were found to be necessary for E2-induced phosphorylation of ERK1/2. This rapid E2 signaling cascade in GnRH neurons was found to require both direct and indirect E2 actions. E2 failed to phosphorylate ERK1/2 and CREB in GnRH neuron-specific estrogen receptor β knockout mice in vivo. Equally, however, a cocktail of tetrodotoxin and γ-aminobutyric acid(A)/glutamate receptor antagonists also blocked E2-induced ERK1/2 phosphorylation in GnRH neurons in wild-type mice in vitro. Together, these observations indicate that E2 acts through calcium/calmodulin-dependent protein kinase type II and protein kinase A to rapidly phosphorylate ERK1/2, which then acts to phosphorylate CREB in adult female GnRH neurons. Intriguingly, these effects of E2 are dependent upon both direct ERβ mechanisms as well as indirect actions mediated by afferent inputs to GnRH neurons.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Lindo Ferguson Building, 270 Great King Street, P.O. Box 913, Dunedin 9054, New Zealand
| | | | | | | | | | | |
Collapse
|
196
|
Chamero P, Leinders-Zufall T, Zufall F. From genes to social communication: molecular sensing by the vomeronasal organ. Trends Neurosci 2012; 35:597-606. [PMID: 22658923 DOI: 10.1016/j.tins.2012.04.011] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 04/27/2012] [Accepted: 04/30/2012] [Indexed: 12/22/2022]
Abstract
The ability to distinguish molecular cues emitted by other individuals is a fundamental feature of social interactions such as finding and identifying a mate, establishing social hierarchies, and initiating interspecies defensive behaviors. In rodents, this ability involves the vomeronasal organ (VNO), a distinct chemoreceptive structure that is part of the olfactory system. Recent insights have led to unprecedented progress in identifying ligand and receptor families underlying vomeronasal recognition, characterizing the behavioral consequences caused by VNO activation, and defining higher neural circuits underlying the initiation of instinctive behaviors such as aggression. Here, we review such findings and discuss future areas for investigation, including large-scale mapping studies, immune system-VNO interactions, in vivo recording of neural activity, and optogenetic alteration of sexual and social behaviors.
Collapse
Affiliation(s)
- Pablo Chamero
- Department of Physiology, University of Saarland School of Medicine, D-66424 Homburg, Germany
| | | | | |
Collapse
|
197
|
Dalfó D, Michaelson D, Hubbard EJA. Sensory regulation of the C. elegans germline through TGF-β-dependent signaling in the niche. Curr Biol 2012; 22:712-9. [PMID: 22483938 DOI: 10.1016/j.cub.2012.02.064] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 12/16/2011] [Accepted: 02/23/2012] [Indexed: 01/16/2023]
Abstract
The proliferation/differentiation balance of stem and progenitor cell populations must respond to the physiological needs of the organism [1, 2]. Mechanisms underlying this plasticity are not well understood. The C. elegans germline provides a tractable system to study the influence of the environment on progenitor cells (stem cells and their proliferative progeny). Germline progenitors accumulate during larval stages to form an adult pool from which gametes are produced. Notch pathway signaling from the distal tip cell (DTC) niche to the germline maintains the progenitor pool [3-5], and the larval germline cell cycle is boosted by insulin/IGF-like receptor signaling [6]. Here we show that, independent of its role in the dauer decision, TGF-β regulates the balance of proliferation versus differentiation in the C. elegans germline in response to sensory cues that report population density and food abundance. Ciliated ASI sensory neurons are required for TGF-β-mediated expansion of the larval germline progenitor pool, and the TGF-β receptor pathway acts in the germline stem cell niche. TGF-β signaling thereby couples germline development to the quality of the environment, providing a novel cellular and molecular mechanism linking sensory experience of the environment to reproduction.
Collapse
Affiliation(s)
- Diana Dalfó
- Developmental Genetics Program, Skirball Institute of Biomolecular Medicine, Helen and Martin Kimmel Center for Stem Cell Biology, and Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | | | |
Collapse
|
198
|
Sabado V, Barraud P, Baker CVH, Streit A. Specification of GnRH-1 neurons by antagonistic FGF and retinoic acid signaling. Dev Biol 2012; 362:254-62. [PMID: 22200593 PMCID: PMC4561506 DOI: 10.1016/j.ydbio.2011.12.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 12/08/2011] [Accepted: 12/09/2011] [Indexed: 11/25/2022]
Abstract
A small population of neuroendocrine cells in the rostral hypothalamus and basal forebrain is the key regulator of vertebrate reproduction. They secrete gonadotropin-releasing hormone (GnRH-1), communicate with many areas of the brain and integrate multiple inputs to control gonad maturation, puberty and sexual behavior. In humans, disruption of the GnRH-1 system leads to hypogonadotropic gonadism and Kallmann syndrome. Unlike other neurons in the central nervous system, GnRH-1 neurons arise in the periphery, however their embryonic origin is controversial, and the molecular mechanisms that control their initial specification are not clear. Here, we provide evidence that in chick GnRH-1 neurons originate in the olfactory placode, where they are specified shortly after olfactory sensory neurons. FGF signaling is required and sufficient to induce GnRH-1 neurons, while retinoic acid represses their formation. Both pathways regulate and antagonize each other and our results suggest that the timing of signaling is critical for normal GnRH-1 neuron formation. While Kallmann's syndrome has generally been attributed to a failure of GnRH-1 neuron migration due to impaired FGF signaling, our findings suggest that in at least some Kallmann patients these neurons may never be specified. In addition, this study highlights the intimate embryonic relationship between GnRH-1 neurons and their targets and modulators in the adult.
Collapse
Affiliation(s)
- Virginie Sabado
- Department of Craniofacial Development, King’s College London, Guy’s Campus, London, SE1 9RT, UK
| | - Perrine Barraud
- Department of Physiology, Development & Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY, UK
| | - Clare V. H. Baker
- Department of Physiology, Development & Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY, UK
| | - Andrea Streit
- Department of Craniofacial Development, King’s College London, Guy’s Campus, London, SE1 9RT, UK
| |
Collapse
|
199
|
Larsen CM, Grattan DR. Prolactin, neurogenesis, and maternal behaviors. Brain Behav Immun 2012; 26:201-9. [PMID: 21820505 DOI: 10.1016/j.bbi.2011.07.233] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 07/04/2011] [Accepted: 07/14/2011] [Indexed: 11/17/2022] Open
Abstract
Elevated prolactin during pregnancy increases neurogenesis in the subventricular zone of the lateral ventricle (SVZ) of the maternal brain. Evidence from our laboratory has shown that low prolactin in early pregnancy, and the consequent suppression of neurogenesis in the SVZ in the adult brain, is associated with increased postpartum anxiety and markedly impaired maternal behavior. Daughters of low prolactin mothers also display increased anxiety and a significant delay in the onset of puberty, which is associated with epigenetic changes in neuronal development (see Fig. 1). This suggests that, in rodents, low prolactin in early pregnancy exerts long-term effects that influence maternal mood postpartum, and offspring development. This mini-review aims to summarize the evidence showing that the prolactin-induced increase in SVZ neurogenesis during pregnancy underlies normal postpartum maternal interactions with pups.
Collapse
Affiliation(s)
- C M Larsen
- Centre for Neuroendocrinology, Department of Anatomy and Structural Biology, University of Otago, Dunedin, New Zealand.
| | | |
Collapse
|
200
|
Ma M. Odor and pheromone sensing via chemoreceptors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 739:93-106. [PMID: 22399397 DOI: 10.1007/978-1-4614-1704-0_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Evolutionally, chemosensation is an ancient but yet enigmatic sense. All organisms ranging from the simplest unicellular form to the most advanced multicellular creature possess the capability to detect chemicals in the surroundings. Conversely, all living things emit some forms of smells, either as communicating signals or as by-products of metabolism. Many species (from worms, insects to mammals) rely on the olfactory systems which express a large number of chemoreceptors to locate food and mates and to avoid danger. Most chemoreceptors expressed in olfactory organs are G-protein coupled receptors (GPCRs) and can be classified into two major categories: odorant receptors (ORs) and pheromone receptors, which principally detect general odors and pheromones, respectively. In vertebrates, these two types of receptors are often expressed in two distinct apparatuses: The main olfactory epithelium (MOE) and the vomeronasal organ (VNO), respectively. Each olfactory sensory neuron (OSN) in the MOE typically expresses one type of OR from a large repertoire. General odors activate ORs and their host OSNs (ranging from narrowly- to broadly-tuned) in a combinatorial manner and the information is sent to the brain via the main olfactory system leading to perception of smells. In contrast, pheromones stimulate relatively narrowly-tuned receptors and their host VNO neurons and the information is sent to the brain via the accessory olfactory system leading to behavioral and endocrinological changes. Recent studies indicate that the functional separation between these two systems is blurred in some cases and there are more subsystems serving chemosensory roles. This chapter focuses on the molecular and cellular mechanisms underlying odor and pheromone sensing in rodents, the best characterized vertebrate models.
Collapse
Affiliation(s)
- Minghong Ma
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|