516 results on '"Median Eminence physiology"'
Search Results
2. Electroacupuncture Promotes the Survival of the Grafted Human MGE Neural Progenitors in Rats with Cerebral Ischemia by Promoting Angiogenesis and Inhibiting Inflammation.
- Author
-
Li J, Chen L, Li D, Lu M, Huang X, Han X, and Chen H
- Subjects
- Animals, Brain Ischemia metabolism, Brain Ischemia pathology, Cell Survival physiology, Cells, Cultured, Embryonic Stem Cells physiology, Embryonic Stem Cells transplantation, Hippocampus cytology, Hippocampus physiology, Humans, Inflammation Mediators metabolism, Male, Maze Learning physiology, Median Eminence cytology, Median Eminence physiology, Neural Stem Cells physiology, Neural Stem Cells transplantation, Rats, Rats, Sprague-Dawley, Brain Ischemia therapy, Electroacupuncture methods, Inflammation Mediators antagonists & inhibitors, Median Eminence transplantation, Neovascularization, Physiologic physiology, Stem Cell Transplantation methods
- Abstract
Stem cells have the potential as a regenerative therapy for cerebral ischemia by improving functional outcomes. However, cell transplantation has some limitations, including a low rate of the grafted cell survival. There is still a major challenge of promoting the harmonious symbiosis between grafted cells and the host. Acupuncture can effectively improve the functional outcome after cerebral ischemia. The present study evaluated the therapeutic effects and explored the mechanism of combined medial ganglionic eminence (MGE) neural progenitors differentiated from human embryonic stem cells (hESCs) with electroacupuncture (EA) in a bilateral common carotid artery occlusion (2VO) rat model. The results showed that EA could promote the survival of the grafted MGE neural progenitors differentiated from hESCs and alleviate learning and memory impairment in rats with cerebral ischemia. This may have partially resulted from inhibited expression of TNF- α and IL-1 β and increased vascular endothelial growth factor (VEGF) expression and blood vessel density in the hippocampus. Our findings indicated that EA could promote the survival of the grafted MGE neural progenitors and enhance transplantation therapy's efficacy by promoting angiogenesis and inhibiting inflammation., Competing Interests: The authors have declared no conflict of interest., (Copyright © 2021 Juan Li et al.)
- Published
- 2021
- Full Text
- View/download PDF
3. Hypothalamic Rax + tanycytes contribute to tissue repair and tumorigenesis upon oncogene activation in mice.
- Author
-
Mu W, Li S, Xu J, Guo X, Wu H, Chen Z, Qiao L, Helfer G, Lu F, Liu C, and Wu QF
- Subjects
- Animals, Carcinogenesis pathology, Cell Self Renewal physiology, Craniopharyngioma chemically induced, Craniopharyngioma genetics, Eye Proteins metabolism, Female, Homeodomain Proteins metabolism, Median Eminence cytology, Mice, Neoplasms, Experimental chemically induced, Neoplasms, Experimental genetics, Proto-Oncogene Proteins B-raf genetics, RNA-Seq, Receptor, IGF Type 1 metabolism, Signal Transduction, Single-Cell Analysis, Transcription Factors metabolism, Craniopharyngioma pathology, Ependymoglial Cells physiology, Median Eminence physiology, Neoplasms, Experimental pathology, Regeneration
- Abstract
Hypothalamic tanycytes in median eminence (ME) are emerging as a crucial cell population that regulates endocrine output, energy balance and the diffusion of blood-born molecules. Tanycytes have recently been considered as potential somatic stem cells in the adult mammalian brain, but their regenerative and tumorigenic capacities are largely unknown. Here we found that Rax+ tanycytes in ME of mice are largely quiescent but quickly enter the cell cycle upon neural injury for self-renewal and regeneration. Mechanistically, Igf1r signaling in tanycytes is required for tissue repair under injury conditions. Furthermore, Braf oncogenic activation is sufficient to transform Rax+ tanycytes into actively dividing tumor cells that eventually develop into a papillary craniopharyngioma-like tumor. Together, these findings uncover the regenerative and tumorigenic potential of tanycytes. Our study offers insights into the properties of tanycytes, which may help to manipulate tanycyte biology for regulating hypothalamic function and investigate the pathogenesis of clinically relevant tumors.
- Published
- 2021
- Full Text
- View/download PDF
4. MCH Neurons Regulate Permeability of the Median Eminence Barrier.
- Author
-
Jiang H, Gallet S, Klemm P, Scholl P, Folz-Donahue K, Altmüller J, Alber J, Heilinger C, Kukat C, Loyens A, Müller-Fielitz H, Sundaram S, Schwaninger M, Prevot V, and Brüning JC
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus physiology, Blood Vessels physiology, Capillaries physiology, Cell Nucleus physiology, Cell Nucleus ultrastructure, Endothelial Cells physiology, Leptin physiology, Median Eminence blood supply, Mice, Primary Cell Culture, Rats, Rats, Sprague-Dawley, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors, Vascular Endothelial Growth Factor A biosynthesis, Cell Membrane Permeability physiology, Hypothalamic Hormones physiology, Median Eminence physiology, Melanins physiology, Neurons physiology, Pituitary Hormones physiology
- Abstract
Melanin-concentrating hormone (MCH)-expressing neurons are key regulators of energy and glucose homeostasis. Here, we demonstrate that they provide dense projections to the median eminence (ME) in close proximity to tanycytes and fenestrated vessels. Chemogenetic activation of MCH neurons as well as optogenetic stimulation of their projections in the ME enhance permeability of the ME by increasing fenestrated vascular loops and enhance leptin action in the arcuate nucleus of the hypothalamus (ARC). Unbiased phosphoRiboTrap-based assessment of cell activation upon chemogenetic MCH neuron activation reveals MCH-neuron-dependent regulation of endothelial cells. MCH neurons express the vascular endothelial growth factor A (VEGFA), and blocking VEGF-R signaling attenuates the leptin-sensitizing effect of MCH neuron activation. Our experiments reveal that MCH neurons directly regulate permeability of the ME barrier, linking the activity of energy state and sleep regulatory neurons to the regulation of hormone accessibility to the ARC., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
5. GABAergic Restriction of Network Dynamics Regulates Interneuron Survival in the Developing Cortex.
- Author
-
Duan ZRS, Che A, Chu P, Modol L, Bollmann Y, Babij R, Fetcho RN, Otsuka T, Fuccillo MV, Liston C, Pisapia DJ, Cossart R, and De Marco García NV
- Subjects
- Animals, Apoptosis physiology, Cell Survival physiology, Excitatory Postsynaptic Potentials physiology, Female, GABAergic Neurons metabolism, Inhibitory Postsynaptic Potentials physiology, Interneurons metabolism, Male, Median Eminence physiology, Membrane Potentials physiology, Mice, Mice, Transgenic, Neural Pathways physiology, Neurogenesis physiology, Parvalbumins metabolism, Pyramidal Cells metabolism, Pyramidal Cells physiology, Somatosensory Cortex growth & development, Somatostatin metabolism, Synaptic Potentials physiology, gamma-Aminobutyric Acid metabolism, GABAergic Neurons physiology, Interneurons physiology, Somatosensory Cortex physiology
- Abstract
During neonatal development, sensory cortices generate spontaneous activity patterns shaped by both sensory experience and intrinsic influences. How these patterns contribute to the assembly of neuronal circuits is not clearly understood. Using longitudinal in vivo calcium imaging in un-anesthetized mouse pups, we show that spatially segregated functional assemblies composed of interneurons and pyramidal cells are prominent in the somatosensory cortex by postnatal day (P) 7. Both reduction of GABA release and synaptic inputs onto pyramidal cells erode the emergence of functional topography, leading to increased network synchrony. This aberrant pattern effectively blocks interneuron apoptosis, causing increased survival of parvalbumin and somatostatin interneurons. Furthermore, the effect of GABA on apoptosis is mediated by inputs from medial ganglionic eminence (MGE)-derived but not caudal ganglionic eminence (CGE)-derived interneurons. These findings indicate that immature MGE interneurons are fundamental for shaping GABA-driven activity patterns that balance the number of interneurons integrating into maturing cortical networks., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
6. Human Pluripotent Stem Cell-Derived Striatal Interneurons: Differentiation and Maturation In Vitro and in the Rat Brain.
- Author
-
Noakes Z, Keefe F, Tamburini C, Kelly CM, Cruz Santos M, Dunnett SB, Errington AC, and Li M
- Subjects
- Animals, Corpus Striatum metabolism, GABAergic Neurons cytology, GABAergic Neurons metabolism, Hippocampus metabolism, Humans, Interneurons metabolism, Median Eminence metabolism, Median Eminence physiology, Neurogenesis physiology, Pluripotent Stem Cells metabolism, RNA, Messenger metabolism, Rats, Cell Differentiation physiology, Corpus Striatum cytology, Hippocampus cytology, Interneurons cytology, Pluripotent Stem Cells cytology
- Abstract
Striatal interneurons are born in the medial and caudal ganglionic eminences (MGE and CGE) and play an important role in human striatal function and dysfunction in Huntington's disease and dystonia. MGE/CGE-like neural progenitors have been generated from human pluripotent stem cells (hPSCs) for studying cortical interneuron development and cell therapy for epilepsy and other neurodevelopmental disorders. Here, we report the capacity of hPSC-derived MGE/CGE-like progenitors to differentiate into functional striatal interneurons. In vitro, these hPSC neuronal derivatives expressed cortical and striatal interneuron markers at the mRNA and protein level and displayed maturing electrophysiological properties. Following transplantation into neonatal rat striatum, progenitors differentiated into striatal interneuron subtypes and were consistently found in the nearby septum and hippocampus. These findings highlight the potential for hPSC-derived striatal interneurons as an invaluable tool in modeling striatal development and function in vitro or as a source of cells for regenerative medicine., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
7. CTCF Governs the Identity and Migration of MGE-Derived Cortical Interneurons.
- Author
-
Elbert A, Vogt D, Watson A, Levy M, Jiang Y, Brûlé E, Rowland ME, Rubenstein J, and Bérubé NG
- Subjects
- Animals, CCCTC-Binding Factor genetics, Cell Count, Cell Movement genetics, Cell Movement physiology, Cerebral Cortex cytology, Female, LIM-Homeodomain Proteins biosynthesis, LIM-Homeodomain Proteins genetics, Male, Median Eminence cytology, Mice, Mice, Inbred C57BL, Neocortex cytology, Neocortex physiology, Nerve Tissue Proteins biosynthesis, Nerve Tissue Proteins genetics, Parvalbumins metabolism, Somatostatin metabolism, Telencephalon cytology, Telencephalon growth & development, Transcription Factors biosynthesis, Transcription Factors genetics, gamma-Aminobutyric Acid physiology, CCCTC-Binding Factor physiology, Cerebral Cortex physiology, Interneurons physiology, Median Eminence physiology
- Abstract
The CCCTC-binding factor (CTCF) is a central regulator of chromatin topology recently linked to neurodevelopmental disorders such as intellectual disability, autism, and schizophrenia. The aim of this study was to identify novel roles of CTCF in the developing mouse brain. We provide evidence that CTCF is required for the expression of the LIM homeodomain factor LHX6 involved in fate determination of cortical interneurons (CINs) that originate in the medial ganglionic eminence (MGE). Conditional Ctcf ablation in the MGE of mice of either sex leads to delayed tangential migration, abnormal distribution of CIN in the neocortex, a marked reduction of CINs expressing parvalbumin and somatostatin (Sst), and an increased number of MGE-derived cells expressing Lhx8 and other markers of basal forebrain projection neurons. Likewise, Ctcf -null MGE cells transplanted into the cortex of wild-type hosts generate fewer Sst-expressing CINs and exhibit lamination defects that are efficiently rescued upon reexpression of LHX6. Collectively, these data indicate that CTCF regulates the dichotomy between Lhx6 and Lhx8 to achieve correct specification and migration of MGE-derived CINs. SIGNIFICANCE STATEMENT This work provides evidence that CCCTC-binding factor (CTCF) controls an early fate decision point in the generation of cortical interneurons mediated at least in part by Lhx6. Importantly, the abnormalities described could reflect early molecular and cellular events that contribute to human neurological disorders previously linked to CTCF, including schizophrenia, autism, and intellectual disability., (Copyright © 2019 the authors 0270-6474/19/390177-16$15.00/0.)
- Published
- 2019
- Full Text
- View/download PDF
8. MGE-derived nNOS + interneurons promote fear acquisition in nNOS -/- mice.
- Author
-
Zhang L, Yuan HJ, Cao B, Kong CC, Yuan F, Li J, Ni HY, Wu HY, Chang L, Liu Y, and Luo CX
- Subjects
- Animals, Behavior, Animal physiology, Cells, Cultured, Dentate Gyrus cytology, Dentate Gyrus physiology, Dentate Gyrus surgery, Interneurons cytology, Interneurons transplantation, Learning physiology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Neural Stem Cells cytology, Neural Stem Cells physiology, Neural Stem Cells transplantation, Nitric Oxide Synthase Type I deficiency, Nitric Oxide Synthase Type I genetics, Telencephalon cytology, Telencephalon embryology, Fear physiology, Interneurons physiology, Median Eminence physiology, Nitric Oxide Synthase Type I physiology
- Abstract
Neuronal nitric oxide synthase (nNOS)
1 , mainly responsible for NO release in central nervous system (CNS)2 , plays a significant role in multiple physiological functions. However, the function of nNOS+ interneurons in fear learning has not been much explored. Here we focused on the medial ganglionic eminences (MGE)3 -derived nNOS+ interneurons in fear learning. To determine the origin of nNOS+ interneurons, we cultured neurons in vitro from MGE, cortex, lateral ganglionic eminence (LGE)4 , caudal ganglionic eminences (CGE)5 and preoptic area (POA)6 . The results showed that MGE contained the most abundant precursors of nNOS+ interneurons. Moreover, donor cells from E12.5 embryos demonstrated the highest positive rate of nNOS+ interneurons compared with other embryonic periods (E11.5, E12, E13, E13.5 and E14). Additionally, these cells from E12.5 embryos showed long axonal and abundant dendritic arbors after 10 days culture, indicating the capability to disperse and integrate in host neural circuits after transplantation. To investigate the role of MGE-derived nNOS+ interneurons in fear learning, donor MGE cells were transplanted into dentate gyrus (DG)7 of nNOS knock-out (nNOS-/- ) or wild-type mice. Results showed that the transplantation of MGE cells promoted the acquisition of nNOS-/- but not the wild-type mice, suggesting the importance of nNOS+ neurons in fear acquisition. Moreover, we transplanted MGE cells from nNOS-/- mice or wild-type mice into DG of the nNOS-/- mice and found that only MGE cells from wild-type mice but not the nNOS-/- mice rescued the deficit in acquisition of the nNOS-/- mice, further confirming the positive role of nNOS+ neurons in fear learning., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
9. Multiple-scale neuroendocrine signals connect brain and pituitary hormone rhythms.
- Author
-
Romanò N, Guillou A, Hodson DJ, Martin AO, and Mollard P
- Subjects
- Action Potentials physiology, Animals, Biological Clocks physiology, Electrochemical Techniques, Female, Mice, Mice, Inbred C57BL, Microelectrodes, Hypothalamus physiology, Median Eminence physiology, Pituitary Gland physiology, Pituitary-Adrenal System physiology, Prolactin metabolism, Ultradian Rhythm physiology
- Abstract
Small assemblies of hypothalamic "parvocellular" neurons release their neuroendocrine signals at the median eminence (ME) to control long-lasting pituitary hormone rhythms essential for homeostasis. How such rapid hypothalamic neurotransmission leads to slowly evolving hormonal signals remains unknown. Here, we show that the temporal organization of dopamine (DA) release events in freely behaving animals relies on a set of characteristic features that are adapted to the dynamic dopaminergic control of pituitary prolactin secretion, a key reproductive hormone. First, locally generated DA release signals are organized over more than four orders of magnitude (0.001 Hz-10 Hz). Second, these DA events are finely tuned within and between frequency domains as building blocks that recur over days to weeks. Third, an integration time window is detected across the ME and consists of high-frequency DA discharges that are coordinated within the minutes range. Thus, a hierarchical combination of time-scaled neuroendocrine signals displays local-global integration to connect brain-pituitary rhythms and pace hormone secretion.
- Published
- 2017
- Full Text
- View/download PDF
10. Embryonic interneurons from the medial, but not the caudal ganglionic eminence trigger ocular dominance plasticity in adult mice.
- Author
-
Isstas M, Teichert M, Bolz J, and Lehmann K
- Subjects
- Animals, Cell Movement, GABAergic Neurons physiology, Male, Mice, Mice, Inbred C57BL, Photic Stimulation, Sensory Deprivation physiology, Visual Perception physiology, Dominance, Ocular, Interneurons physiology, Median Eminence physiology, Neuronal Plasticity, Visual Cortex physiology
- Abstract
The maturation of cortical inhibition provided by parvalbumin-containing basket cells derived from the medial ganglionic eminence (MGE) is a key event in starting the enhanced visual cortical plasticity during the critical period. Although it is generally assumed that a further increase in inhibition closes the critical period again, it was recently shown that embryonic interneurons derived from the MGE can induce an additional, artificial critical period when injected into the visual cortex of young mice. It has, however, remained open whether this effect was indeed specific for MGE-derived cells, and whether critical period-like plasticity could also be induced in fully adult animals. To clarify these issues, we injected explants from either the MGE or the caudal ganglionic eminence (CGE) into the visual cortices of fully adult mice, and performed monocular deprivation 33 days later for 4 days. Animals implanted with MGE cells, but not with CGE cells, showed marked ocular dominance plasticity. Immunohistochemistry confirmed that the injected cells from both sources migrated far in the host cortex, that most developed into neurons producing GABA, and that only cells from the MGE expressed parvalbumin. Thus, our results confirm that the plasticity-inducing effect of embryonic interneurons is specific for cells from the MGE, and is independent of the host animal's age.
- Published
- 2017
- Full Text
- View/download PDF
11. Extended Production of Cortical Interneurons into the Third Trimester of Human Gestation.
- Author
-
Arshad A, Vose LR, Vinukonda G, Hu F, Yoshikawa K, Csiszar A, Brumberg JC, and Ballabh P
- Subjects
- Brain metabolism, Cell Count, Cerebral Cortex embryology, Cerebral Cortex metabolism, Cerebral Cortex physiology, Female, GABAergic Neurons metabolism, Gestational Age, Homeodomain Proteins metabolism, Humans, Interneurons metabolism, Lateral Ventricles embryology, Lateral Ventricles metabolism, Lateral Ventricles physiology, Male, Median Eminence embryology, Median Eminence physiology, Neural Stem Cells metabolism, Neurogenesis, Nuclear Proteins metabolism, Pregnancy, Pregnancy Trimester, Third, Thyroid Nuclear Factor 1, Transcription Factors metabolism, Brain embryology, Brain physiology, Fetal Development, GABAergic Neurons physiology, Interneurons physiology, Neural Stem Cells physiology
- Abstract
In humans, the developmental origins of interneurons in the third trimester of pregnancy and the timing of completion of interneuron neurogenesis have remained unknown. Here, we show that the total and cycling Nkx2.1(+)and Dlx2(+)interneuron progenitors as well as Sox2(+)precursor cells were higher in density in the medial ganglionic eminence (MGE) compared with the lateral ganglionic eminence and cortical ventricular/subventricular zone (VZ/SVZ) of 16-35 gw subjects. The proliferation of these progenitors reduced as a function of gestational age, almost terminating by 35 gw. Proliferating Dlx2(+)cells were higher in density in the caudal ganglionic eminence (CGE) compared with the MGE, and persisted beyond 35 gw. Consistent with these findings, Sox2, Nkx2.1, Dlx2, and Mash1 protein levels were higher in the ganglionic eminences relative to the cortical VZ/SVZ. The density of gamma-aminobutyric acid-positive (GABA(+)) interneurons was higher in the cortical VZ/SVZ relative to MGE, but Nkx2.1 or Dlx2-expressing GABA(+)cells were more dense in the MGE compared with the cortical VZ/SVZ. The data suggest that the MGE and CGE are the primary source of cortical interneurons. Moreover, their generation continues nearly to the end of pregnancy, which may predispose premature infants to neurobehavioral disorders., (© The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
12. Viral-mediated Labeling and Transplantation of Medial Ganglionic Eminence (MGE) Cells for In Vivo Studies.
- Author
-
Vogt D, Wu PR, Sorrells SF, Arnold C, Alvarez-Buylla A, and Rubenstein JL
- Subjects
- Animals, Female, GABAergic Neurons cytology, GABAergic Neurons physiology, GABAergic Neurons virology, HEK293 Cells, Humans, Interneurons cytology, Interneurons transplantation, Lentivirus genetics, Median Eminence cytology, Median Eminence transplantation, Mice, Neural Stem Cells cytology, Pregnancy, Signal Transduction, Transduction, Genetic, Cell Transplantation methods, GABAergic Neurons transplantation, Interneurons physiology, Interneurons virology, Median Eminence physiology, Median Eminence virology
- Abstract
GABAergic cortical interneurons, derived from the embryonic medial and caudal ganglionic eminences (MGE and CGE), are functionally and morphologically diverse. Inroads have been made in understanding the roles of distinct cortical interneuron subgroups, however, there are still many mechanisms to be worked out that may contribute to the development and maturation of different types of GABAergic cells. Moreover, altered GABAergic signaling may contribute to phenotypes of autism, schizophrenia and epilepsy. Specific Cre-driver lines have begun to parcel out the functions of unique interneuron subgroups. Despite the advances in mouse models, it is often difficult to efficiently study GABAergic cortical interneuron progenitors with molecular approaches in vivo. One important technique used to study the cell autonomous programming of these cells is transplantation of MGE cells into host cortices. These transplanted cells migrate extensively, differentiate, and functionally integrate. In addition, MGE cells can be efficiently transduced with lentivirus immediately prior to transplantation, allowing for a multitude of molecular approaches. Here we detail a protocol to efficiently transduce MGE cells before transplantation for in vivo analysis, using available Cre-driver lines and Cre-dependent expression vectors. This approach is advantageous because it combines precise genetic manipulation with the ability of these cells to disperse after transplantation, permitting greater cell-type specific resolution in vivo.
- Published
- 2015
- Full Text
- View/download PDF
13. Semaphorin7A regulates neuroglial plasticity in the adult hypothalamic median eminence.
- Author
-
Parkash J, Messina A, Langlet F, Cimino I, Loyens A, Mazur D, Gallet S, Balland E, Malone SA, Pralong F, Cagnoni G, Schellino R, De Marchis S, Mazzone M, Pasterkamp RJ, Tamagnone L, Prevot V, and Giacobini P
- Subjects
- Analysis of Variance, Animals, Antigens, CD administration & dosage, Blotting, Western, Enzyme-Linked Immunosorbent Assay, Estradiol analogs & derivatives, Female, Flow Cytometry, Fluorescent Antibody Technique, Image Processing, Computer-Assisted, Immunohistochemistry, Mice, Neuronal Plasticity drug effects, Ovariectomy, Progesterone, Rats, Rats, Sprague-Dawley, Real-Time Polymerase Chain Reaction, Semaphorins administration & dosage, Antigens, CD pharmacology, Median Eminence physiology, Neuroglia metabolism, Neuronal Plasticity physiology, Semaphorins pharmacology
- Abstract
Reproductive competence in mammals depends on the projection of gonadotropin-releasing hormone (GnRH) neurons to the hypothalamic median eminence (ME) and the timely release of GnRH into the hypothalamic-pituitary-gonadal axis. In adult rodents, GnRH neurons and the specialized glial cells named tanycytes periodically undergo cytoskeletal plasticity. However, the mechanisms that regulate this plasticity are still largely unknown. We demonstrate that Semaphorin7A, expressed by tanycytes, plays a dual role, inducing the retraction of GnRH terminals and promoting their ensheathment by tanycytic end feet via the receptors PlexinC1 and Itgb1, respectively. Moreover, Semaphorin7A expression is regulated during the oestrous cycle by the fluctuating levels of gonadal steroids. Genetic invalidation of Semaphorin7A receptors in mice induces neuronal and glial rearrangements in the ME and abolishes normal oestrous cyclicity and fertility. These results show a role for Semaphorin7A signalling in mediating periodic neuroglial remodelling in the adult ME during the ovarian cycle.
- Published
- 2015
- Full Text
- View/download PDF
14. Hypothalamus as an endocrine organ.
- Author
-
Clarke IJ
- Subjects
- Animals, Feedback, Physiological physiology, Humans, Hypothalamic Hormones metabolism, Hypothalamic Hormones physiology, Hypothalamus cytology, Median Eminence physiology, Neuroendocrine Cells physiology, Neurosecretory Systems physiology, Neurotransmitter Agents metabolism, Pituitary Gland, Anterior physiology, Hypothalamus physiology
- Abstract
The endocrine hypothalamus constitutes those cells which project to the median eminence and secrete neurohormones into the hypophysial portal blood to act on cells of the anterior pituitary gland. The entire endocrine system is controlled by these peptides. In turn, the hypothalamic neuroendocrine cells are regulated by feedback signals from the endocrine glands and other circulating factors. The neuroendocrine cells are found in specific regions of the hypothalamus and are regulated by afferents from higher brain centers. Integrated function is clearly complex and the networks between and amongst the neuroendocrine cells allows fine control to achieve homeostasis. The entry of hormones and other factors into the brain, either via the cerebrospinal fluid or through fenestrated capillaries (in the basal hypothalamus) is important because it influences the extent to which feedback regulation may be imposed. Recent evidence of the passage of factors from the pars tuberalis and the median eminence casts a new layer in our understanding of neuroendocrine regulation. The function of neuroendocrine cells and the means by which pulsatile secretion is achieved is best understood for the close relationship between gonadotropin releasing hormone and luteinizing hormone, which is reviewed in detail. The secretion of other neurohormones is less rigid, so the relationship between hypothalamic secretion and the relevant pituitary hormones is more complex., (© 2015 American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
15. Long-lasting anxiolytic effect of neural precursor cells freshly prepared but not neurosphere-derived cell transplantation in newborn rats.
- Author
-
Romariz SA, Paiva Dde S, Valente MF, Barnabé GF, Frussa-Filho R, Barbosa-Silva RC, Calcagnotto ME, and Longo BM
- Subjects
- Animals, Animals, Newborn, Anxiety physiopathology, Calbindin 2 metabolism, Cell Culture Techniques, Cell Movement physiology, Cerebral Cortex physiopathology, Embryonic Stem Cells physiology, Exploratory Behavior physiology, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Hippocampus physiopathology, Interneurons physiology, Male, Median Eminence embryology, Median Eminence physiology, Neural Stem Cells physiology, Neurogenesis physiology, Neuropeptide Y metabolism, Parvalbumins metabolism, Rats, Sprague-Dawley, Rats, Transgenic, Anxiety therapy, Cell Transplantation methods, Embryonic Stem Cells transplantation, Median Eminence transplantation, Neural Stem Cells transplantation
- Abstract
Background: The GABAergic system plays an important role in modulating levels of anxiety. When transplanted into the brain, precursor cells from the medial ganglionic eminence (MGE) have the ability to differentiate into GABAergic interneurons and modify the inhibitory tone in the host brain. Currently, two methods have been reported for obtaining MGE precursor cells for transplantation: fresh and neurosphere dissociated cells. Here, we investigated the effects generated by transplantation of the two types of cell preparations on anxiety behavior in rats., Results: We transplanted freshly dissociated or neurosphere dissociated cells into the neonate brain of male rats on postnatal (PN) day 2-3. At early adulthood (PN 62-63), transplanted animals were tested in the Elevated Plus Maze (EPM). To verify the differentiation and migration pattern of the transplanted cells in vitro and in vivo, we performed immunohistochemistry for GFP and several interneuron-specific markers: neuropeptide Y (NPY), parvalbumin (PV) and calretinin (CR). Cells from both types of preparations expressed these interneuronal markers. However, an anxiolytic effect on behavior in the EPM was observed in animals that received the MGE-derived freshly dissociated cells but not in those that received the neurosphere dissociated cells., Conclusion: Our results suggest a long-lasting anxiolytic effect of transplanted freshly dissociated cells that reinforces the inhibitory function of the GABAergic neuronal circuitry in the hippocampus related to anxiety-like behavior in rats.
- Published
- 2014
- Full Text
- View/download PDF
16. Activation of neurokinin 3 receptors stimulates GnRH release in a location-dependent but kisspeptin-independent manner in adult mice.
- Author
-
Gaskins GT, Glanowska KM, and Moenter SM
- Subjects
- Animals, Female, Gene Expression Regulation drug effects, Gonadotropin-Releasing Hormone genetics, Kisspeptins genetics, Male, Median Eminence physiology, Mice, Mice, Knockout, Peptide Fragments pharmacology, Receptors, Neurokinin-3 agonists, Receptors, Neurokinin-3 genetics, Substance P analogs & derivatives, Substance P pharmacology, Gonadotropin-Releasing Hormone metabolism, Kisspeptins metabolism, Receptors, Neurokinin-3 metabolism
- Abstract
GnRH neurons form the final common pathway for the central control of reproduction. GnRH release occurs from terminals in the external layer of the median eminence (ME) for neuroendocrine control of the pituitary, and near GnRH-GnRH fiber appositions within the preoptic area (POA). Whether or not control of GnRH secretion by neuromodulators is different in these 2 areas is unknown. Mutations in neurokinin B (NKB) or the neurokinin-3 receptor (NK3R) are linked to hypogonadotropic hypogonadism in humans, suggesting that NKB may regulate GnRH secretion. Using fast scan cyclic voltammetry through carbon-fiber microelectrodes, we examined real-time GnRH release in response to the NK3R agonist senktide in the ME and POA. Coronal brain slices were acutely prepared from adult gonad-intact GnRH-green fluorescent protein male mice, and carbon-fiber microelectrodes were placed either within green fluorescent protein-positive terminal fields of the ME or near GnRH-GnRH fiber appositions in the POA. Senktide induced GnRH release consistently in the ME but not the POA, indicating that GnRH release is differentially regulated by NKB in a location-dependent manner. Senktide also induced GnRH secretion in the ME of kisspeptin-knockout (Kiss1 knockout) mice. Interestingly, release amplitude was lower compared with wild-type mice. These data indicate regulation of GnRH release by NK3R agonists is site specific and suggest that kisspeptin is not a required mediator between NK3R activation and GnRH secretion in the ME. This information will be useful for informing future models of afferent regulation of GnRH release.
- Published
- 2013
- Full Text
- View/download PDF
17. Tanycyte-like cells form a blood-cerebrospinal fluid barrier in the circumventricular organs of the mouse brain.
- Author
-
Langlet F, Mullier A, Bouret SG, Prevot V, and Dehouck B
- Subjects
- Animals, Antibodies chemistry, Area Postrema physiology, Blood-Brain Barrier metabolism, Brain cytology, Cell Membrane Permeability, Cerebral Ventricles, Ependymoglial Cells metabolism, Immunohistochemistry, Male, Median Eminence cytology, Mice, Mice, Inbred C57BL, Permeability, Subcommissural Organ physiology, Subfornical Organ physiology, Tight Junction Proteins metabolism, Blood-Brain Barrier physiology, Brain physiology, Cerebrospinal Fluid physiology, Ependymoglial Cells physiology, Median Eminence physiology
- Abstract
Tanycytes are highly specialized ependymal cells that form a blood-cerebrospinal fluid (CSF) barrier at the level of the median eminence (ME), a circumventricular organ (CVO) located in the tuberal region of the hypothalamus. This ependymal layer harbors well-organized tight junctions, a hallmark of central nervous system barriers that is lacking in the fenestrated portal vessels of the ME. The displacement of barrier properties from the vascular to the ventricular side allows the diffusion of blood-borne molecules into the parenchyma of the ME while tanycyte tight junctions control their diffusion into the CSF, thus maintaining brain homeostasis. In the present work, we combined immunohistochemical and permeability studies to investigate the presence of tanycyte barriers along the ventricular walls of other brain CVOs. Our data indicate that, unlike cuboidal ependymal cells, ependymal cells bordering the CVOs possess long processes that project into the parenchyma of the CVOs to reach the fenestrated capillary network. Remarkably, these tanycyte-like cells display well-organized tight junctions around their cell bodies. Consistent with these observations, permeability studies show that this ependymal layer acts as a diffusion barrier. Together, our results suggest that tanycytes are a characteristic feature of all CVOs and yield potential new insights into their involvement in regulating the exchange between the blood, the brain, and the CSF within these "brain windows.", (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2013
- Full Text
- View/download PDF
18. Rapid sensing of circulating ghrelin by hypothalamic appetite-modifying neurons.
- Author
-
Schaeffer M, Langlet F, Lafont C, Molino F, Hodson DJ, Roux T, Lamarque L, Verdié P, Bourrier E, Dehouck B, Banères JL, Martinez J, Méry PF, Marie J, Trinquet E, Fehrentz JA, Prévot V, and Mollard P
- Subjects
- Animals, Blood-Brain Barrier physiology, Capillary Permeability, Eating physiology, Fasting physiology, Hypothalamus blood supply, Hypothalamus cytology, Male, Median Eminence blood supply, Median Eminence cytology, Median Eminence physiology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microscopy, Fluorescence, Multiphoton, Models, Neurological, Neurons physiology, Appetite Regulation physiology, Ghrelin blood, Hypothalamus physiology
- Abstract
To maintain homeostasis, hypothalamic neurons in the arcuate nucleus must dynamically sense and integrate a multitude of peripheral signals. Blood-borne molecules must therefore be able to circumvent the tightly sealed vasculature of the blood-brain barrier to rapidly access their target neurons. However, how information encoded by circulating appetite-modifying hormones is conveyed to central hypothalamic neurons remains largely unexplored. Using in vivo multiphoton microscopy together with fluorescently labeled ligands, we demonstrate that circulating ghrelin, a versatile regulator of energy expenditure and feeding behavior, rapidly binds neurons in the vicinity of fenestrated capillaries, and that the number of labeled cell bodies varies with feeding status. Thus, by virtue of its vascular connections, the hypothalamus is able to directly sense peripheral signals, modifying energy status accordingly.
- Published
- 2013
- Full Text
- View/download PDF
19. GABAergic interneuron migration and the evolution of the neocortex.
- Author
-
Tanaka DH and Nakajima K
- Subjects
- Animals, Chickens physiology, Embryo, Mammalian physiology, GABAergic Neurons physiology, Interneurons physiology, Median Eminence cytology, Median Eminence physiology, Median Eminence transplantation, Neocortex cytology, Phylogeny, Rodentia physiology, Species Specificity, Turtles physiology, Biological Evolution, Cell Movement, GABAergic Neurons cytology, Interneurons cytology, Neocortex physiology
- Abstract
A neocortex is present in all mammals but is not present in other classes of vertebrates, and the neocortex is extremely elaborate in humans. Changes in excitatory projection neurons and their progenitors within the developing dorsal pallium in the most recent common ancestor of mammals are thought to have been involved in the evolution of the neocortex. Our recent findings suggest that changes in the migratory ability of inhibitory interneurons derived from outside the neocortex may also have been involved in the evolution of the neocortex. In this article we review the literature on the migratory profile of inhibitory interneurons in several different species and the literature on comparisons between the intrinsic migratory ability of interneurons derived from different species. Finally, we propose a hypothesis about the mammalian-specific evolution of the migratory ability of interneurons and its potential contribution to the establishment of a functional neocortex., (© 2012 The Authors Development, Growth & Differentiation © 2012 Japanese Society of Developmental Biologists.)
- Published
- 2012
- Full Text
- View/download PDF
20. Rac1-dependent cell cycle exit of MGE precursors and GABAergic interneuron migration to the cortex.
- Author
-
Vidaki M, Tivodar S, Doulgeraki K, Tybulewicz V, Kessaris N, Pachnis V, and Karagogeos D
- Subjects
- Animals, Cerebral Cortex cytology, Cerebral Cortex pathology, Female, G1 Phase genetics, Interneurons cytology, Interneurons pathology, Median Eminence cytology, Median Eminence pathology, Mice, Mice, Knockout, Neural Stem Cells cytology, Neural Stem Cells pathology, Neuropeptides deficiency, Neuropeptides genetics, Pregnancy, Primary Cell Culture, rac GTP-Binding Proteins deficiency, rac GTP-Binding Proteins genetics, rac1 GTP-Binding Protein, Cell Cycle Checkpoints genetics, Cell Movement genetics, Cerebral Cortex physiology, Interneurons physiology, Median Eminence physiology, Neural Stem Cells physiology, Neuropeptides physiology, rac GTP-Binding Proteins physiology
- Abstract
Cortical γ-aminobutyric acid (GABA)ergic interneurons are characterized by extraordinary neurochemical and functional diversity. Although recent studies have uncovered some of the molecular components underlying interneuron development, including the cellular and molecular mechanisms guiding their migration to the cortex, the intracellular components involved are still unknown. Rac1, a member of the Rac subfamily of Rho-GTPases, has been implicated in various cellular processes such as cell cycle dynamics, axonogenesis, and migration. In this study, we have addressed the specific role of Rac1 in interneuron progenitors originating in the medial ganglionic eminence, via Cre/loxP technology. We show that ablation of Rac1 from Nkx2.1-positive progenitors, results in a migratory impairment. As a consequence, only half of GABAergic interneurons are found in the postnatal cortex. The rest remain aggregated in the ventral telencephalon and show morphological defects in their growing processes in vitro. Ablation of Rac1 from postmitotic progenitors does not result in similar defects, thus underlying a novel cell autonomous and stage-specific requirement for Rac1 activity, within proliferating progenitors of cortical interneurons. Rac1 is necessary for their transition from G1 to S phase, at least in part by regulating cyclin D levels and retinoblastoma protein phosphorylation.
- Published
- 2012
- Full Text
- View/download PDF
21. The function of the adrenocortical axis in permanent middle cerebral artery occlusion: effect of glucocorticoids on the neurological outcome.
- Author
-
Weidenfeld J, Leker RR, Gai N, Teichner A, Bener D, and Ovadia H
- Subjects
- Adrenalectomy, Adrenocorticotropic Hormone blood, Animals, Anti-Inflammatory Agents therapeutic use, Brain Ischemia complications, Corticosterone blood, Corticotropin-Releasing Hormone metabolism, Dexamethasone therapeutic use, Dinoprostone metabolism, Hypothalamo-Hypophyseal System physiology, Hypothalamus metabolism, Infarction, Middle Cerebral Artery etiology, Infarction, Middle Cerebral Artery physiopathology, Lameness, Animal etiology, Lameness, Animal pathology, Male, Median Eminence physiology, Peptide Fragments metabolism, Rats, Rats, Inbred SHR, Receptors, Glucocorticoid drug effects, Receptors, Glucocorticoid physiology, Stroke etiology, Treatment Outcome, Adrenal Cortex physiology, Brain Ischemia physiopathology, Glucocorticoids physiology, Glucocorticoids therapeutic use, Infarction, Middle Cerebral Artery drug therapy, Stroke physiopathology
- Abstract
We characterized the effect of acute ischemic stroke on the activation of the hypothalamic-pituitary-adrenal (HPA) axis and evaluated the role of glucocorticoids (GC) in the clinical outcome following ischemic stroke. Male spontaneous hypertensive rats underwent permanent middle cerebral artery occlusion (PMCAO) and developed a cortical infarct. At 4h post-PMCAO or sham operation, serum levels of ACTH and corticosterone (CS) were elevated 5 and 4 fold respectively as compared to controls and then returned to basal levels at 24h post surgery. In these experimental groups we found also a significant depletion of median eminence (ME)-CRH(41). In adrenalectomized (Adx) rats that underwent PMCAO the degree of motor disability and infarct volume was similar to that of intact rats. Administration of dexamethasone (Dex) to Adx-PMCAO rats significantly improved the motor disability and decreased the infarct volume. However, in sham-Adx with PMCAO, Dex had no effect on these two parameters. In rats with PMCAO or sham-PMCAO, brain production of PGE(2) was significantly increased. This effect was further enhanced in Adx-PMCAO rats and significantly inhibited by Dex. In conclusion, activation of the HPA axis following PMCAO is due to stress induced by surgery. This activation is mediated by hypothalamic CRH(41). Absence of endogenous GC or administration of Dex in naïve rats does not alter motor and pathological parameters in the acute stage following PMCAO. In contrast, administration of Dex significantly improved the outcome following cerebral ischemia in Adx rats which may be due to increased glucocorticoid receptors. Brain production of PGE(2) does not play an important role in the pathophysiology of the acute phase of cerebral ischemia., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
22. Metabolic sensing and the brain: who, what, where, and how?
- Author
-
Levin BE, Magnan C, Dunn-Meynell A, and Le Foll C
- Subjects
- Animals, Astrocytes physiology, Humans, Median Eminence physiology, Brain physiology, Energy Metabolism, Homeostasis, Sensory Receptor Cells physiology
- Abstract
Unique subpopulations of specialized metabolic sensing neurons reside in a distributed network throughout the brain and respond to alterations in ambient levels of various metabolic substrates by altering their activity. Variations in local brain substrate levels reflect their transport across the blood- and cerebrospinal-brain barriers as well as local production by astrocytes. There are a number of mechanisms by which such metabolic sensing neurons alter their activity in response to changes in substrate levels, but it is clear that these neurons cannot be considered in isolation. They are heavily dependent on astrocyte and probably tanycyte metabolism and function but also respond to hormones (e.g. leptin and insulin) and cytokines that cross the blood-brain barrier from the periphery as well as hard-wired neural inputs from metabolic sensors in peripheral sites such as the hepatic portal vein, gastrointestinal tract, and carotid body. Thus, these specialized neurons are capable of monitoring and integrating multiple signals from the periphery as a means of regulating peripheral energy homeostasis.
- Published
- 2011
- Full Text
- View/download PDF
23. Loss of COUP-TFI alters the balance between caudal ganglionic eminence- and medial ganglionic eminence-derived cortical interneurons and results in resistance to epilepsy.
- Author
-
Lodato S, Tomassy GS, De Leonibus E, Uzcategui YG, Andolfi G, Armentano M, Touzot A, Gaztelu JM, Arlotta P, Menendez de la Prida L, and Studer M
- Subjects
- Animals, Antimetabolites, Bromodeoxyuridine, Cell Proliferation, Cerebral Cortex cytology, Convulsants pharmacology, Drug Resistance genetics, Electroencephalography, Electrophysiological Phenomena, Epilepsy chemically induced, Immunohistochemistry, In Situ Hybridization, Median Eminence cytology, Mice, Mice, Inbred C57BL, Mice, Knockout, Nerve Net cytology, Nerve Net physiology, Receptors, GABA-A genetics, Receptors, GABA-A physiology, Receptors, GABA-B genetics, Receptors, GABA-B physiology, Telencephalon cytology, Telencephalon physiology, gamma-Aminobutyric Acid physiology, COUP Transcription Factor I genetics, COUP Transcription Factor I physiology, Cerebral Cortex physiology, Epilepsy genetics, Epilepsy physiopathology, Interneurons physiology, Median Eminence physiology
- Abstract
In rodents, cortical interneurons originate from the medial ganglionic eminence (MGE) and caudal ganglionic eminence (CGE) according to precise temporal schedules. The mechanisms controlling the specification of CGE-derived interneurons and their role in cortical circuitry are still unknown. Here, we show that COUP-TFI expression becomes restricted to the dorsal MGE and CGE at embryonic day 13.5 in the basal telencephalon. Conditional loss of function of COUP-TFI in subventricular precursors and postmitotic cells leads to a decrease of late-born, CGE-derived, VIP (vasoactive intestinal peptide)- and CR (calretinin)-expressing bipolar cortical neurons, compensated by the concurrent increase of early-born MGE-derived, PV (parvalbumin)-expressing interneurons. Strikingly, COUP-TFI mutants are more resistant to pharmacologically induced seizures, a phenotype that is dependent on GABAergic signaling. Together, our data indicate that COUP-TFI controls the delicate balance between MGE- and CGE-derived cortical interneurons by regulating intermediate progenitor divisions and ultimately affecting the activity of the cortical inhibitory circuitry.
- Published
- 2011
- Full Text
- View/download PDF
24. Expression of kv4.3 voltage-gated potassium channels in rat gonadotrophin-releasing hormone (GnRH) neurons during the estrous cycle.
- Author
-
Arroyo A, Kim BS, Biehl A, Yeh J, and Bett GC
- Subjects
- Animals, Blotting, Western, Female, Gene Expression physiology, Ion Channel Gating physiology, Median Eminence cytology, Median Eminence physiology, Preoptic Area cytology, Preoptic Area physiology, Presynaptic Terminals physiology, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, Estrous Cycle physiology, Gonadotropin-Releasing Hormone physiology, Neurons physiology, Shal Potassium Channels genetics, Shal Potassium Channels metabolism
- Abstract
Regular and timely electrical activity of gonadotrophin-releasing hormone (GnRH) neurons accompanies the pulsatile release of GnRH that plays a central role in regulating fertility. Although transient outward A-type currents (I(A)) have been electrophysiologically identified in GnRH neurons, the molecular identity of the channels that underlie these currents are unknown. Several families of voltage-gated potassium channels can underlie I(A). However, the biophysical properties of I(A) described in previous electrophysiological studies are strongly characteristic of members of the Kv4 family of voltage-gated channels. We, therefore, sought to determine the presence of Kv4 channels in GnRH neurons. We used reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis to determine whether Kv4 messenger RNA (mRNA) and protein are present in the rat medial preoptic area (MPOA) and median eminence (ME). We used double-label immunohistochemistry to determine whether Kv4 colocalized with GnRH cell bodies in the MPOA and GnRH axons in the ME. Kv4.3 channels co-localized with GnRH in the MPOA but not in the ME. Neither Kv4.2 nor Kv4.1 co-localized with GnRH in either the MPOA or the ME. The electrical activity of GnRH neurons changes dramatically during the estrous cycle. We, therefore, studied the change in Kv4.3 expression in GnRH neurons during the estrous cycle. In the estrus phase, 58.05% of GnRH neurons expressed Kv4.3 compared to 74.48% in diestrus-proestrus rats (P < .05). Our data suggest that Kv4.3 is the major molecular component of I(A) in GnRH neurons, and furthermore that the expression of Kv4.3 changes significantly during the rat estrous cycle.
- Published
- 2011
- Full Text
- View/download PDF
25. A novel developmental role for kisspeptin in the growth of gonadotrophin-releasing hormone neurites to the median eminence in the mouse.
- Author
-
Fiorini Z and Jasoni CL
- Subjects
- Animals, Collagen metabolism, Dose-Response Relationship, Drug, Embryo, Mammalian anatomy & histology, Female, Humans, Kisspeptins, Median Eminence physiology, Mice, Mice, Transgenic, Neurites drug effects, Neurites ultrastructure, Pregnancy, RNA, Messenger metabolism, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Receptors, Kisspeptin-1, Signal Transduction physiology, Tissue Culture Techniques, Tumor Suppressor Proteins genetics, Type C Phospholipases antagonists & inhibitors, Gonadotropin-Releasing Hormone metabolism, Median Eminence cytology, Neurites physiology, Tumor Suppressor Proteins pharmacology
- Abstract
The puberty- and fertility-regulating neuropeptide kisspeptin (KISS1) exerts dramatic effects on the physiology of adult gonadotrophin-releasing hormone (GnRH) neurones as a master regulator of mammalian reproduction. Given the action of KISS1 directly on adult GnRH neurones, and that KISS1 activates a signal transduction cascade involved in neurite growth in other neurones, we investigated whether KISS1 may play a role in the normal growth of GnRH neurites to the median eminence. A reverse transcription-polymerase chain reaction demonstrated the expression of Kiss1 mRNA in the embryonic mediobasal hypothalamus, the target region for GnRH neurite termination, as early as embryonic day 13.5 (E13.5), a time when the first GnRH neurites are arriving. Complementary expression of the mRNA encoding the KISS1 receptor, Kiss1r, in the preoptic area (POA) at E13.5 was also observed, suggesting that POA-resident GnRH neurones can respond to KISS1 from an early age. To examine the effects of KISS1 on GnRH neurite growth in isolation, E15.5 POA explants, containing GnRH neurones actively extending neurites, were grown in three-dimensional collagen gels. In the presence of KISS1 (1 μm), both the number and length of GnRH neurites were increased significantly compared to controls without KISS1. The effects of KISS1 on GnRH neurite growth could be inhibited by pretreatment with the phospholipase C inhibitor U73122 (50 μm), indicating that embryonic and adult GnRH neurones respond to KISS1 with the same intracellular signalling pathway. KISS1 provided in a concentration gradient from a fixed source had no effect on GnRH neurite growth, indicating that KISS1 does not function as a long-range chemoattractant. Taken together, these results identify KISS1 as a stimulator of GnRH neurite growth, and suggest that it influences GnRH neurites at close-range to innervate the median eminence. These data add a novel developmental role to the repertoire of the functions of KISS1 in mammalian reproduction., (© 2010 The Authors. Journal of Neuroendocrinology © 2010 Blackwell Publishing Ltd.)
- Published
- 2010
- Full Text
- View/download PDF
26. Seasonal changes in hypothalamic gonadotropin-releasing hormone-I immunoreactivity in relation with testicular volume in adult male free-living European starlings (Sturnus vulgaris).
- Author
-
Pintér O and Péczely P
- Subjects
- Animals, Immunohistochemistry, Male, Median Eminence physiology, Preoptic Area physiology, Reproduction physiology, Seasons, Starlings anatomy & histology, Testis anatomy & histology, Gonadotropin-Releasing Hormone physiology, Hypothalamus physiology, Starlings physiology
- Abstract
Birds from the temperate and cold zones show annual sexual activity accompanied by gonadal changes and fluctuation in their brain gonadotropin-releasing hormone (GnRH) levels. However, most of the studies were done on captive birds where the constant environment can profoundly modify periodical changes. Therefore our aim was to reveal annual variations of hypothalamic and gonadal changes in male, free-living European starlings (Sturnus vulgaris) captured directly from their natural environment. We analyzed hypothalamic GnRH-I immunoreactivity and testes volume. Four key time points of the active reproductive cycle and the photorefractory phase were studied. GnRH-I immunoreactivity was analyzed in the preoptic area (POA) and the median eminence (ME). Photorefractory birds (August) with regressed gonads had the lowest level of GnRH-I immunoreactivity compared to other birds from the active reproductive phases. These results suggest that parallel with the gonadal volume GnRH-I undergoes seasonal changes in adult male free-living European starlings.
- Published
- 2010
- Full Text
- View/download PDF
27. A mammalian neural tissue opsin (Opsin 5) is a deep brain photoreceptor in birds.
- Author
-
Nakane Y, Ikegami K, Ono H, Yamamoto N, Yoshida S, Hirunagi K, Ebihara S, Kubo Y, and Yoshimura T
- Subjects
- Amino Acid Sequence, Animals, Avian Proteins genetics, Brain anatomy & histology, Coturnix anatomy & histology, Coturnix genetics, Female, In Vitro Techniques, Male, Median Eminence anatomy & histology, Median Eminence physiology, Models, Neurological, Molecular Sequence Data, Nerve Tissue Proteins genetics, Neural Pathways anatomy & histology, Neural Pathways physiology, Oocytes metabolism, Opsins genetics, Paraventricular Hypothalamic Nucleus anatomy & histology, Paraventricular Hypothalamic Nucleus physiology, Photic Stimulation, Recombinant Proteins genetics, Recombinant Proteins metabolism, Testis growth & development, Xenopus laevis, Avian Proteins physiology, Brain physiology, Coturnix physiology, Nerve Tissue Proteins physiology, Opsins physiology, Photoreceptor Cells, Vertebrate physiology
- Abstract
It has been known for many decades that nonmammalian vertebrates detect light by deep brain photoreceptors that lie outside the retina and pineal organ to regulate seasonal cycle of reproduction. However, the identity of these photoreceptors has so far remained unclear. Here we report that Opsin 5 is a deep brain photoreceptive molecule in the quail brain. Expression analysis of members of the opsin superfamily identified as Opsin 5 (OPN5; also known as Gpr136, Neuropsin, PGR12, and TMEM13) mRNA in the paraventricular organ (PVO), an area long believed to be capable of phototransduction. Immunohistochemistry identified Opsin 5 in neurons that contact the cerebrospinal fluid in the PVO, as well as fibers extending to the external zone of the median eminence adjacent to the pars tuberalis of the pituitary gland, which translates photoperiodic information into neuroendocrine responses. Heterologous expression of Opsin 5 in Xenopus oocytes resulted in light-dependent activation of membrane currents, the action spectrum of which showed peak sensitivity (lambda(max)) at approximately 420 nm. We also found that short-wavelength light, i.e., between UV-B and blue light, induced photoperiodic responses in eye-patched, pinealectomized quail. Thus, Opsin 5 appears to be one of the deep brain photoreceptive molecules that regulates seasonal reproduction in birds.
- Published
- 2010
- Full Text
- View/download PDF
28. The hypothalamic median eminence and its role in reproductive aging.
- Author
-
Yin W and Gore AC
- Subjects
- Aging metabolism, Animals, Gonadotropin-Releasing Hormone metabolism, Humans, Hypothalamus metabolism, Hypothalamus physiology, Median Eminence metabolism, Neuroglia metabolism, Neuroglia physiology, Aging physiology, Median Eminence physiology, Reproduction physiology
- Abstract
The median eminence at the base of the hypothalamus serves as an interface between the neural and peripheral endocrine systems. It releases hypothalamic-releasing hormones into the portal capillary bed for transport to the anterior pituitary, which provides further signals to target endocrine systems. Of specific relevance to reproduction, a group of about 1000 neurons in mammals release the gonadotropin-releasing hormone (GnRH) peptide from neuroterminals in the median eminence. During the life cycle, there are dramatic changes in reproductive demands, and we focus this review on how GnRH terminals in the median eminence change during reproductive senescence. We discuss morphological and functional properties of the median eminence, and how relationships among GnRH terminals and their microenvironment of nerve terminals, glial cells, and the portal capillary vasculature determine the ability of GnRH peptide to be secreted and to reach its target in the anterior pituitary gland.
- Published
- 2010
- Full Text
- View/download PDF
29. Influence of age and 17beta-estradiol on kisspeptin, neurokinin B, and prodynorphin gene expression in the arcuate-median eminence of female rhesus macaques.
- Author
-
Eghlidi DH, Haley GE, Noriega NC, Kohama SG, and Urbanski HF
- Subjects
- Age Factors, Aging metabolism, Aging physiology, Animals, Arcuate Nucleus of Hypothalamus drug effects, Arcuate Nucleus of Hypothalamus physiology, Enkephalins metabolism, Female, Gene Expression drug effects, Genes, Tumor Suppressor drug effects, Macaca mulatta, Median Eminence drug effects, Median Eminence physiology, Neurokinin B metabolism, Ovariectomy, Protein Precursors metabolism, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Time Factors, Aging genetics, Arcuate Nucleus of Hypothalamus metabolism, Enkephalins genetics, Estradiol pharmacology, Median Eminence metabolism, Neurokinin B genetics, Protein Precursors genetics
- Abstract
The neuropeptides kisspeptin, neurokinin B, and dynorphin A (collectively abbreviated as KNDy) are, respectively, encoded by KiSS-1, NKB, and PDYN and are coexpressed by neurons of the hypothalamic arcuate nucleus (ARC). Here, using quantitative real-time PCR, we examined age-related changes in the expression of genes encoding KNDy and associated receptors G protein-coupled receptor 54 (encoded by GPR54), neurokinin 3 receptor (encoded by NK3), and kappa-opioid receptor (encoded by KOR), in the female rhesus macaque ARC-median eminence (ARC-ME). Expression of KiSS-1 and NKB was highly elevated in old perimenopausal compared with young or middle-aged premenopausal animals. To test whether these age-related changes could be attributed to perimenopausal loss of sex steroids, we then examined KNDy, GPR54, NK3, and KOR expression changes in response to ovariectomy (OVX) and exposure to 17beta-estradiol (E(2)). Short-term (7 months) OVX (with or without 1 month of estrogen replacement) failed to modulate the expression of any of the KNDy-related genes. In contrast, long-term ( approximately 4 yr) OVX significantly increased KiSS-1 and NKB expression, and this was reversed by E(2) administration. Finally, we examined the expression of KNDy-related genes in young adult females during the early follicular, late follicular, or midluteal phases of their menstrual cycle but found no difference. Together, the results suggest that short-term alterations in circulating E(2) levels, such as those occurring during the menstrual cycle, may have little effect on the ARC-ME expression of KNDy and associated receptors. Nevertheless, they clearly demonstrate that loss of ovarian steroid negative feedback that occurs during perimenopause plays a major role in modulating the activity of KNDy circuits of the aging primate ARC-ME.
- Published
- 2010
- Full Text
- View/download PDF
30. Role of estradiol in the dynamic control of tanycyte plasticity mediated by vascular endothelial cells in the median eminence.
- Author
-
de Seranno S, d'Anglemont de Tassigny X, Estrella C, Loyens A, Kasparov S, Leroy D, Ojeda SR, Beauvillain JC, and Prevot V
- Subjects
- Analysis of Variance, Animals, Blotting, Western, Cell Communication drug effects, Cell Communication physiology, Cell Culture Techniques, Cell Shape drug effects, Cells, Cultured, Dinoprostone pharmacology, Endothelial Cells drug effects, Ependyma drug effects, Estradiol pharmacology, Hypothalamo-Hypophyseal System physiology, Neuroglia drug effects, Nitric Oxide Synthase Type III physiology, Prostaglandin-Endoperoxide Synthases physiology, Rats, Rats, Sprague-Dawley, Cell Shape physiology, Endothelial Cells physiology, Ependyma physiology, Estradiol physiology, Median Eminence physiology, Neuroglia physiology
- Abstract
In the ever-changing physiological context of the neuroendocrine brain, the mechanisms by which cellular events involving neurons, astroglia, and vascular cells are coordinated to bring forth the appropriate neuronal signaling is not yet known but is amenable to examination. In the median eminence of the hypothalamus, endothelial cells are key players in the plasticity of tanycytes (specialized astroglia) and neuroendocrine synapse efficacy. Here we report that estradiol acts on both purified endothelial cells and isolated tanycytes to trigger endothelial-to-glial communication that leads to a sudden and massive retraction of tanycyte processes. The blockade of endothelial nitric oxide synthase by in vitro adenoviral-mediated gene transfer of a dominant-negative form of endothelial nitric oxide synthase abrogates the estradiol-induced tanycyte plasticity mediated by endothelial cells. In parallel, increases in prostaglandin-E(2) (PGE(2)) due to changes in cyclooxygenase (COX)-1 and COX-2 expression induced by the exposure of tanycytes to estradiol promote acute tanycyte plasticity. We also demonstrate by electron microscopy that the administration of PGE(2) to median eminence explants induces rapid neuroglial plasticity at the neurovascular junction of neurons that release GnRH (the neuropeptide controlling reproduction). Conversely, preventing local PGE(2) synthesis in the median eminence of adult female rats with the COX inhibitor indomethacin impairs the ovarian cycle, a process that requires a pulsatile, coordinated delivery of GnRH into the hypothalamo-hypophyseal portal system. Taken together, our findings show that estradiol controls the dialog between endothelial cells and astroglia to regulate neuroglial plasticity in the neuroendocrine brain.
- Published
- 2010
- Full Text
- View/download PDF
31. Interactions between neurotensin and GnRH neurons in the positive feedback control of GnRH/LH secretion in the mouse.
- Author
-
Dungan Lemko HM, Naderi R, Adjan V, Jennes LH, Navarro VM, Clifton DK, and Steiner RA
- Subjects
- Animals, Cell Communication physiology, Estradiol pharmacology, Estrogens pharmacology, Feedback, Physiological drug effects, Female, Genes, fos physiology, Hypothalamus, Middle cytology, Hypothalamus, Middle physiology, Immunohistochemistry, Injections, Intraventricular, Kisspeptins, Median Eminence cytology, Median Eminence physiology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neurotensin genetics, Ovariectomy, Preoptic Area cytology, Preoptic Area physiology, RNA, Messenger metabolism, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Feedback, Physiological physiology, Gonadotropin-Releasing Hormone metabolism, Luteinizing Hormone metabolism, Neurons metabolism, Neurotensin metabolism
- Abstract
In female mammals, increased ovarian estradiol (E(2)) secretion triggers GnRH release from neurons in the basal forebrain, which drives LH secretion from the pituitary and subsequently induces ovulation. However, the neural circuits that activate this preovulatory GnRH/LH surge remain unidentified. Neurotensin is expressed in neurons of the anteroventral periventricular nucleus (AVPV), a region thought to be critical for generating the preovulatory GnRH/LH surge. E(2) induces neurotensin (Nts) gene expression in this region, and blockade of neurotensin signaling reduces the LH surge in the rat. We postulated that neurotensin signaling plays a similar role in generating the E(2)-induced GnRH/LH surge in mice. We used in situ hybridization (ISH) to determine whether E(2) induces Nts expression in the mouse and found evidence to support this proposition. Next, we determined that the neurotensin receptor (Ntsr2) is present in many GnRH-expressing neurons. Since the kisspeptin gene (Kiss1) is expressed in the AVPV and is responsive to E(2), we predicted that some neurons in this region express both Kiss1 and Nts; however, by double-label ISH, we observed no coexpression of the two mRNAs. We also postulated that Nts mRNA expression would increase in parallel with the E(2)-induced LH surge and that the central (icv) administration of neurotensin would stimulate LH secretion and activation of GnRH neurons but found no evidence to support either of these hypotheses. Together, these findings suggest that, although neurotensin neurons in the AVPV are targets for regulation by E(2), neurotensin does not appear to play a direct role in generating the GnRH/LH surge in the mouse.
- Published
- 2010
- Full Text
- View/download PDF
32. Neuropeptide W has cell phenotype-specific effects on the excitability of different subpopulations of paraventricular nucleus neurones.
- Author
-
Price CJ, Samson WK, and Ferguson AV
- Subjects
- Action Potentials, Animals, Corticotropin-Releasing Hormone metabolism, Male, Median Eminence physiology, Medulla Oblongata physiology, Membrane Potentials physiology, Neural Inhibition physiology, Neural Pathways physiology, Oxytocin metabolism, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Spinal Cord physiology, Thyrotropin-Releasing Hormone metabolism, Vasopressins metabolism, Neurons physiology, Neuropeptides metabolism, Paraventricular Hypothalamic Nucleus physiology
- Abstract
The administration of the neuropeptide W (NPW) and neuropeptide B (NPB) in rodents has been shown to influence the activity of a variety of autonomic and neuroendocrine systems. The paraventricular nucleus (PVN) is a major autonomic and neuroendocrine integration site in the hypothalamus, and neurones within this nucleus express the receptor for these ligands, NPB/W receptor 1 (NPBWR1). In the present study, we used whole cell patch clamp recordings coupled with single-cell reverse transcriptase-polymerase chain reaction to examine the effects of neuropeptide W-23 (NPW-23) on the excitability of identified PVN neurones. Oxytocin, vasopressin and thyrotrophin-releasing hormone neurones were all found to be responsive to 10 nm NPW-23, although both depolarising and hyperpolarising effects were observed in each of these cell groups. By contrast, corticotrophin-releasing hormone cells were unaffected. Further subdivision of chemically phenotyped cell groups into magnocellular, neuroendocrine or pre-autonomic neurones, using their electrophysiological fingerprints, revealed that neurones projecting to medullary and spinal targets were predominantly inhibited by NPW-23, whereas those that projected to median eminence or neural lobe showed almost equivalent numbers of depolarising and hyperpolarising cells. The demonstration of particular phenotypic populations of PVN neurones showing NPW-induced effects on excitability reinforces the importance of the NPB/NPW neuropeptide system as a regulator of autonomic function.
- Published
- 2009
- Full Text
- View/download PDF
33. A riot of rhythms: neuronal and glial circadian oscillators in the mediobasal hypothalamus.
- Author
-
Guilding C, Hughes AT, Brown TM, Namvar S, and Piggins HD
- Subjects
- Action Potentials drug effects, Animals, Arcuate Nucleus of Hypothalamus drug effects, Arcuate Nucleus of Hypothalamus physiology, Biological Clocks drug effects, Circadian Rhythm drug effects, Colforsin pharmacology, Dietary Fats administration & dosage, Dietary Fats pharmacology, Feeding Behavior drug effects, Food Deprivation, Hypothalamus drug effects, Luciferases metabolism, Luminescent Measurements, Male, Median Eminence drug effects, Median Eminence physiology, Mice, Neuroglia drug effects, Neurons drug effects, Organ Specificity drug effects, Period Circadian Proteins metabolism, Sodium Channels metabolism, Suprachiasmatic Nucleus drug effects, Suprachiasmatic Nucleus physiology, Tetrodotoxin pharmacology, Biological Clocks physiology, Circadian Rhythm physiology, Hypothalamus physiology, Neuroglia metabolism, Neurons metabolism
- Abstract
Background: In mammals, the synchronized activity of cell autonomous clocks in the suprachiasmatic nuclei (SCN) enables this structure to function as the master circadian clock, coordinating daily rhythms in physiology and behavior. However, the dominance of this clock has been challenged by the observations that metabolic duress can over-ride SCN controlled rhythms, and that clock genes are expressed in many brain areas, including those implicated in the regulation of appetite and feeding. The recent development of mice in which clock gene/protein activity is reported by bioluminescent constructs (luciferase or luc) now enables us to track molecular oscillations in numerous tissues ex vivo. Consequently we determined both clock activities and responsiveness to metabolic perturbations of cells and tissues within the mediobasal hypothalamus (MBH), a site pivotal for optimal internal homeostatic regulation., Results: Here we demonstrate endogenous circadian rhythms of PER2::LUC expression in discrete subdivisions of the arcuate (Arc) and dorsomedial nuclei (DMH). Rhythms resolved to single cells did not maintain long-term synchrony with one-another, leading to a damping of oscillations at both cell and tissue levels. Complementary electrophysiology recordings revealed rhythms in neuronal activity in the Arc and DMH. Further, PER2::LUC rhythms were detected in the ependymal layer of the third ventricle and in the median eminence/pars tuberalis (ME/PT). A high-fat diet had no effect on the molecular oscillations in the MBH, whereas food deprivation resulted in an altered phase in the ME/PT., Conclusion: Our results provide the first single cell resolution of endogenous circadian rhythms in clock gene expression in any intact tissue outside the SCN, reveal the cellular basis for tissue level damping in extra-SCN oscillators and demonstrate that an oscillator in the ME/PT is responsive to changes in metabolism.
- Published
- 2009
- Full Text
- View/download PDF
34. Neurochemistry and plasticity of the median eminence and neural pituitary lobe in relation to background adaptation of Xenopus laevis.
- Author
-
van Wijk DC and Roubos EW
- Subjects
- Animals, Adaptation, Biological physiology, Median Eminence physiology, Neuronal Plasticity, Neurons chemistry, Pituitary Gland physiology, Xenopus laevis physiology
- Abstract
Using immunocytochemistry and morphometry we have supported our hypothesis that magnocellular neurons in the preoptic area of the brain of Xenopus laevis release identical sets of neuropeptides containing not only the previously identified vasotocin, mesotocin, corticotropin-releasing factor, thyrotropin-releasing hormone, brain-derived neurotrophic factor, urocortin 1, and pituitary adenylate cyclase-activating peptide but also mesotocin and met-enkephalin from both neurohemal areas in the pituitary neural lobe and in the median eminence. We also show that the external zone of the median eminence is plastic, depending in size on the state of background illumination.
- Published
- 2009
- Full Text
- View/download PDF
35. Neuroendocrine proopiomelanocortin neurons are excited by hypocretin/orexin.
- Author
-
Acuna-Goycolea C and van den Pol AN
- Subjects
- Action Potentials physiology, Animals, Arcuate Nucleus of Hypothalamus physiology, Evans Blue metabolism, Green Fluorescent Proteins genetics, Median Eminence physiology, Mice, Mice, Transgenic, Neurons drug effects, Neurosecretory Systems metabolism, Orexins, Protein Transport genetics, Intracellular Signaling Peptides and Proteins physiology, Neurons physiology, Neuropeptides physiology, Neurosecretory Systems physiology, Pro-Opiomelanocortin physiology
- Abstract
Hypocretin/orexin, produced by a group of neurons in the lateral hypothalamus/perifornical area, enhances cognitive arousal and also may play a crucial role in modulating the neuroendocrine system. How hypocretin modulates the endocrine system remains an open question. Hypocretin cells innervate the mediobasal hypothalamus where they can potentially influence the activity of specific cell populations within the arcuate nucleus. Here, we examine whether hypocretin modulates the median eminence-projecting proopiomelanocortin (POMC) neurons identified by selective green fluorescent protein expression and antidromic stimulation or retrograde Evans blue dye tracing in transgenic mice. We find that POMC neurons, in general, and, in addition, those that project their axons to the median eminence, were robustly activated by hypocretin in a dose-dependent manner. These excitatory actions included a threefold increase in spike frequency and direct membrane depolarization of up to 22 mV (mean, 17.9+/-7.2 mV). Direct postsynaptic depolarization was decreased at more positive membrane potentials, inhibited by the sodium-calcium exchanger antagonist KB-R7943, and reduced by lowering the bath temperature, or by buffering the postsynaptic calcium with BAPTA, suggesting that the primary mechanism for hypocretin-mediated excitation is the activation of the sodium-calcium exchanger. Hypocretin also enhanced excitatory inputs to POMC cells via a presynaptic mechanism and indirectly increased the release of GABA onto these cells in a spike-dependent manner. However, these synaptic actions were not necessary to cause postsynaptic membrane depolarization and spiking. Thus, in contrast to previous suggestions that hypocretin inhibited POMC cells, our results demonstrate robust direct excitation of POMC neurons by hypocretin.
- Published
- 2009
- Full Text
- View/download PDF
36. ICV vs. VMH injection of leptin: comparative effects on hypothalamic gene expression.
- Author
-
Ambati S, Duan J, Choi YH, Hartzell DL, Della-Fera MA, and Baile CA
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus physiology, Body Weight drug effects, Cachexia genetics, Eating drug effects, Eating physiology, Hypothalamus drug effects, Inflammation genetics, Injections, Injections, Intraventricular, Leptin blood, Male, Median Eminence physiology, Organ Size drug effects, RNA, Messenger biosynthesis, RNA, Messenger genetics, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, Gene Expression drug effects, Hypothalamus metabolism, Leptin administration & dosage, Leptin pharmacology, Ventromedial Hypothalamic Nucleus physiology
- Abstract
Leptin regulates feeding behavior and body weight by binding to its receptors localized in specific areas of the hypothalamus. Leptin injected twice daily for 4 days either into the right ventromedial hypothalamus (VMH) or into the right lateral cerebral ventricle (ICV) and using Real-Time Taqman RT-PCR, mRNA expression levels of selected genes in the arcuate nucleus-median eminence (ARC-ME) complex were quantitatively measured. Expression of selected genes from the ipsi- vs. contralateral VMH areas in rats injected with leptin into the VMH was also compared. VMH injections of leptin increased ARC-ME mRNAs of proopiomelanocortin (POMC), 27.3% (p<0.05); gamma-aminobutyric acid A receptor (GABRD), 89.3% (p<0.01); and thyrotropin-releasing hormone (TRH), 57.7% (p<0.01); and decreased janus kinase 2 (JAK2), 44.4% (p<0.001); suppressor of cytokine signaling 3 (SOCS3), 86.6% (p<0.001); signal transducer and activator of transcription 3 (STAT3), 46.8% (p<0.01); tyrosine hydroxylase (TH), 51.1% (p<0.001); prostaglandin E synthase (PTGES), 96.5% (p<0.001); tumor necrosis factor-alpha (TNF-alpha), 47% (p<0.01); and secretin, 55.4% (p<0.001). Only GABRD, 76.6% (p<0.01) and SCT, 64.9% (p<0.01) were up-regulated in the hypothalamic ARC-ME of rats with ICV leptin injections. VMH injections of leptin induced identical reductions in expression levels of CART, SOCS3, PTGES, and TNF-alpha in both VMH areas; except TH mRNA, whose expression was lowered ipsilaterally. Food intake, body and fat pad weights and serum insulin and leptin were also decreased in rats given leptin through VMH. This study suggests that leptin either unilateral exposure through VMH or bilateral exposure through ICV injections induces divergent ARC-ME gene profiles.
- Published
- 2009
- Full Text
- View/download PDF
37. [André Calas, the original path of a neuroendocrinologist].
- Author
-
Tixier-Vidal A
- Subjects
- Animals, Ducks, France, History, 20th Century, History, 21st Century, Median Eminence physiology, Neuronal Plasticity, Neuropeptides physiology, Rats, Universities history, Neuroendocrinology history
- Abstract
This talk, given as an introduction to a symposium organised to honor André Calas, calls forth his personality, recalls the major events in his career and summarizes the evolution of his research.
- Published
- 2009
- Full Text
- View/download PDF
38. Alterations in RFamide-related peptide expression are coordinated with the preovulatory luteinizing hormone surge.
- Author
-
Gibson EM, Humber SA, Jain S, Williams WP 3rd, Zhao S, Bentley GE, Tsutsui K, and Kriegsfeld LJ
- Subjects
- Animals, Cricetinae, Dorsomedial Hypothalamic Nucleus cytology, Dorsomedial Hypothalamic Nucleus physiology, Estrogens metabolism, Estrogens pharmacology, Female, Gonadotropin-Releasing Hormone metabolism, Lighting, Median Eminence cytology, Median Eminence physiology, Mesocricetus, Microscopy, Fluorescence, Neural Pathways, Photoperiod, Pituitary Gland cytology, Pituitary Gland physiology, Suprachiasmatic Nucleus cytology, Suprachiasmatic Nucleus physiology, Circadian Rhythm physiology, Follicular Phase physiology, Luteinizing Hormone metabolism, Neuropeptides metabolism
- Abstract
The preovulatory LH surge is triggered when the circadian pacemaker, the bilateral suprachiasmatic nucleus (SCN), stimulates the GnRH system in the presence of high estrogen concentrations (positive feedback). Importantly, during the remainder of the estrous cycle, estradiol inhibits LH release via negative feedback. We have recently documented the presence of a novel mammalian RFamide-related peptide (RFRP), a putative gonadotropin-inhibitory hormone (GnIH), that presumably acts upstream of GnRH to modulate the negative feedback effects of estrogen. The present series of studies used female Syrian hamsters to examine the possibility that, in addition to driving the LH surge positively, the SCN concomitantly coordinates the removal of steroid-mediated RFRP inhibition of the gonadotropic axis to permit the surge. We found that the SCN forms close appositions with RFRP cells, suggesting the possibility for direct temporal control of RFRP activity. During the time of the LH surge, immediate-early gene expression is reduced in RFRP cells, and this temporal regulation is estrogen dependent. To determine whether projections from the SCN regulate the timed reduction in activation of the RFRP system, we exploited the phenomenon of splitting. In split animals in which the SCN are active in antiphase, activation of the RFRP system is asymmetrical. Importantly, this asymmetry is opposite to the state of the GnRH system. Together, these findings point to novel circadian control of the RFRP system and potential participation in the circuitry controlling ovulatory function.
- Published
- 2008
- Full Text
- View/download PDF
39. Comprehensive spatiotemporal transcriptomic analyses of the ganglionic eminences demonstrate the uniqueness of its caudal subdivision.
- Author
-
Willi-Monnerat S, Migliavacca E, Surdez D, Delorenzi M, Luthi-Carter R, and Terskikh AV
- Subjects
- Animals, Female, Pregnancy, Rats, Rats, Sprague-Dawley, Time Factors, Cerebral Cortex embryology, Cerebral Cortex physiology, Gene Expression Profiling methods, Median Eminence embryology, Median Eminence physiology
- Abstract
The elucidation of mechanisms underlying telencephalic neural development has been limited by the lack of knowledge regarding the molecular and cellular aspects of the ganglionic eminence (GE), an embryonic structure that supplies the brain with diverse sets of GABAergic neurons. Here, we report a comprehensive transcriptomic analysis of this structure including its medial (MGE), lateral (LGE) and caudal (CGE) subdivisions and its temporal dynamics in 12.5 to 16 day-old rat embryos. Surprisingly, comparison across subdivisions showed that CGE gene expression was the most unique providing unbiased genetic evidence for its differentiation from MGE and LGE. The molecular signature of the CGE comprised a large set of genes, including Rwdd3, Cyp26b1, Nr2f2, Egr3, Cpta1, Slit3, and Hod, of which several encode cell signaling and migration molecules such as WNT5A, DOCK9, VSNL1 and PRG1. Temporal analysis of the MGE revealed differential expression of unique sets of cell specification and migration genes, with early expression of Hes1, Lhx2, Ctgf and Mdk, and late enrichment of Olfm3, SerpinE2 and Wdr44. These GE profiles reveal new candidate regulators of spatiotemporally governed GABAergic neuronogenesis.
- Published
- 2008
- Full Text
- View/download PDF
40. Upregulated expression of neuropeptide Y in hypothalamic-pituitary system of rats by chronic dexamethasone administration.
- Author
-
Konno J, Yoshida S, Ina A, Ohmomo H, Shutoh F, Nogami H, and Hisano S
- Subjects
- Agouti-Related Protein metabolism, Animals, Body Weight drug effects, Corticotropin-Releasing Hormone metabolism, Immunohistochemistry, In Situ Hybridization, Male, Median Eminence drug effects, Median Eminence physiology, Neuropeptide Y metabolism, Neurophysins metabolism, Pituitary Gland drug effects, Pituitary Gland physiology, RNA, Messenger metabolism, Rats, Rats, Wistar, Up-Regulation drug effects, Up-Regulation physiology, Dexamethasone pharmacology, Glucocorticoids pharmacology, Hypothalamo-Hypophyseal System drug effects, Hypothalamo-Hypophyseal System physiology, Neuropeptide Y genetics
- Abstract
To study the effect of adrenal steroids on neuropeptide Y (NPY) synthesis in the hypothalamic-pituitary system, we examined NPY expression in rats treated with dexamethasone (a synthetic glucocorticoid) by in situ hybridization and immunohistochemistry. Rats were injected daily with dexamethasone (0.2mg/100g/day for 10 days, sc) or sesame oil (vehicle control), or non-injected (intact control). Relative staining area for corticotropin-releasing hormone or neurophysin II, a vasopressin carrier protein, was increased in the external zone of the median eminence in vehicle control, but was equivalent to that of intact control in the dexamethasone-injected group. Density of NPY-stained fiber varicosities was drastically increased in the external, but not the internal, zone of dexamethasone-injected group, coinciding with the increased NPY hybridization signal level in the arcuate nucleus. Dual-labeling experiments revealed no colocalization of NPY with hypophysiotropic or other peptides examined in single fibers of the median eminence. In the dexamethasone-injected group, expressions of NPY mRNA and peptide were detectable in a few pituitary cells, with some being corticotropes. These results suggest that NPY plays hormonal roles in the hypothalamic-pituitary-adrenal axis.
- Published
- 2008
- Full Text
- View/download PDF
41. Gonadotropin-releasing hormone neuron requirements for puberty, ovulation, and fertility.
- Author
-
Herbison AE, Porteous R, Pape JR, Mora JM, and Hurst PR
- Subjects
- Animals, Cell Count, Estrous Cycle physiology, Female, Gonadotropin-Releasing Hormone genetics, Infertility, Female pathology, Luteinizing Hormone blood, Median Eminence pathology, Median Eminence physiology, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Transgenic, Neurons pathology, Neurons physiology, Ovariectomy, Preoptic Area pathology, Preoptic Area physiology, Fertility physiology, Gonadotropin-Releasing Hormone metabolism, Infertility, Female physiopathology, Ovulation physiology, Sexual Maturation physiology
- Abstract
The absolute requirement for reproduction implies that the hypothalamo-pituitary-gonadal axis, controlling fertility, is an evolutionary robust mechanism. The GnRH neurons of the hypothalamus represent the key cell type within the body dictating fertility. However, the level of functional redundancy within the GnRH neuron population is unknown. As a result of a fortuitous transgene insertion event, GNR23 mice exhibit a marked allele-dependent reduction in GnRH neuron number within their brain. Wild-type mice have approximately 600 GnRH neurons, compared with approximately 200 (34%) and approximately 70 (12%) in GNR23(+/-) and GNR23(-/-) mice, respectively. Using these mice, we examined the minimal GnRH neuron requirements for fertility. Male GNR23(-/-) mice exhibited normal fertility. In contrast, female GNR23(-/-) mice were markedly subfertile, failing to produce normal litters, have estrous cycles, or ovulate. The failure of ovulation resulted from an inability of the few existing GnRH neurons to generate the LH surge. This was not the case, however, for the first cycle at puberty that appeared normal. Together, these observations demonstrate that 12% of the GnRH neuron population is sufficient for pulsatile gonadotropin secretion and puberty onset, whereas between 12 and 34% are required for cyclical control in adult female mice. This indicates that substantial redundancy exists within the GnRH neuronal population and suggests that the great majority of GnRH neurons must be dysfunctional before fertility is affected.
- Published
- 2008
- Full Text
- View/download PDF
42. Multiple signaling pathways involved in the effect of endothelin type B receptor in rat median eminence.
- Author
-
Mathison Y, del Garrido MR, and Israel A
- Subjects
- Animals, Calcium metabolism, Cyclic GMP metabolism, Endothelin B Receptor Antagonists, Endothelin-1 pharmacology, Endothelin-3 pharmacology, Endothelins pharmacology, Enzyme Inhibitors pharmacology, Male, NG-Nitroarginine Methyl Ester pharmacology, Neomycin pharmacology, Nitric Oxide physiology, Nitric Oxide Synthase physiology, Oligopeptides pharmacology, Peptide Fragments pharmacology, Piperidines pharmacology, Protein Synthesis Inhibitors pharmacology, Rats, Rats, Sprague-Dawley, Arcuate Nucleus of Hypothalamus physiology, Median Eminence physiology, Phosphatidylinositols metabolism, Receptor, Endothelin B physiology, Signal Transduction physiology
- Abstract
We assessed the possible link between endothelin receptor mediated phosphoinositide breakdown and NO/cGMP signaling pathways in rat arcuate nucleus-median eminence fragments (AN-ME), brain structures known to contain a rich plexus of nitric oxide synthase (NOS)-containing neurons and fibers, together with densely arranged endothelin ETB-receptors-like immunoreactive fibres. Our data show that ET-1, ET-3 and the ETB-receptors agonist, IRL 1620, increased inositol monophosphate (InsP1) accumulation, NOS activity and cGMP formation, in a similar degree. The stimulatory effect of ETs on InsP1 accumulation and cGMP formation was inhibited by the phospholipase C (PLC) inhibitor, neomycin, and the absence of extracellular calcium, suggesting that calcium is involved in endothelin receptor-induced PLC activation. The L-arginine analog, L-NAME, inhibited ET-1 or IRL1620-stimulated cGMP formation. The ETA receptor antagonists BQ 123, did not alter, while the ETB receptor antagonists BQ788 inhibited ETs-induced increase in the PI metabolism, NOS activity and cGMP generation. Our data indicate that in AN-ME, ETB receptor signals through receptor-mediated calcium dependent-stimulation of phosphoinositide breakdown and activation of NOS/cGMP signaling pathway.
- Published
- 2007
- Full Text
- View/download PDF
43. Inhibition of metastin (kisspeptin-54)-GPR54 signaling in the arcuate nucleus-median eminence region during lactation in rats.
- Author
-
Yamada S, Uenoyama Y, Kinoshita M, Iwata K, Takase K, Matsui H, Adachi S, Inoue K, Maeda KI, and Tsukamura H
- Subjects
- Animals, Animals, Suckling, Arcuate Nucleus of Hypothalamus cytology, Female, Immunohistochemistry, In Situ Hybridization, Injections, Intraventricular, Kisspeptins, Luteinizing Hormone blood, Luteinizing Hormone metabolism, Median Eminence cytology, Neurons physiology, Ovulation Inhibition physiology, Proteins genetics, Proteins pharmacology, RNA, Messenger metabolism, Rats, Rats, Wistar, Receptors, G-Protein-Coupled genetics, Receptors, Kisspeptin-1, Signal Transduction physiology, Third Ventricle, Arcuate Nucleus of Hypothalamus physiology, Lactation physiology, Median Eminence physiology, Proteins metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Follicular development and ovulation are suppressed during lactation in various mammalian species, mainly due to the suppression of pulsatile GnRH/LH secretion. Metastin (kisspeptin-54), a KiSS-1 gene product, is an endogenous ligand for GPR54, a G-protein-coupled receptor, and suggested to play a critical role in regulating the gonadal axis. The present study therefore aims to determine whether metastin (kisspeptin-54)-GPR54 signaling in discrete brain areas is inhibited by the suckling stimulus that causes suppression of LH secretion in lactating rats. Quantitative RT-PCR revealed that the KiSS-1 mRNA level was significantly lower in the arcuate nucleus (ARC)-median eminence region in lactating ovariectomized (OVX) and estrogen-treated OVX rats than in nonlactating controls. KiSS-1 mRNA in the anteroventral periventricular nucleus was kept at a low level in both lactating and nonlactating rats despite estrogen treatment. GPR54 mRNA levels were significantly lower in lactating than nonlactating rats in the anteroventral periventricular nucleus, but the levels in lactating mothers of the preoptic area and ARC-median eminence were comparable with nonlactating controls. Although KiSS-1 mRNA-expressing cells or metastin (kisspeptin-54) immunoreactivities were densely located in the ARC of nonlactating controls, few were found in the ARC of lactating OVX animals. Various doses of metastin (kisspeptin-54) (0.02, 0.2, and 2 nmol) injected into the third ventricle caused a significant increase in LH secretion in both lactating and nonlactating OVX rats, suggesting that lactating rats are responsive to metastin (kisspeptin-54) stimulus. Thus, the present study demonstrated that KiSS-1 mRNA/metastin (kisspeptin-54) expression is inhibited in the ARC by the suckling stimulus, suggesting that the inhibition is most probably involved in suppressing LH secretion in lactating rats.
- Published
- 2007
- Full Text
- View/download PDF
44. Molecular anatomy of the brain endothelial barrier: an overview of the distributional features.
- Author
-
Ueno M
- Subjects
- ATP Binding Cassette Transporter, Subfamily B physiology, ATP Binding Cassette Transporter, Subfamily B, Member 1 physiology, Adherens Junctions physiology, Animals, Cadherins physiology, Endothelium, Vascular cytology, Median Eminence physiology, Mice, Pineal Gland physiology, Pituitary Gland, Posterior physiology, Tight Junctions physiology, Blood-Brain Barrier physiology, Blood-Brain Barrier ultrastructure, Cerebral Ventricles physiology, Endothelium, Vascular physiology
- Abstract
The blood-brain barrier (BBB) impedes the influx of intravascular compounds from the blood to the brain. The elements composing the BBB are endothelial cells, pericytes and the end-feet of astrocytes. Among them, the endothelial cell barrier line is the most critical for preventing toxic substances from entering the brain. In this review, we focus on the ultrastructural distribution of important components in the intracellular junction and cytoplasm of brain endothelial cells. The ultrastructural distribution of tight junction-specific integral membrane proteins such as occludin, junctional adhesion molecules, claudin, peripheral zonula occludens protein-1 (ZO-1), adherens junction-specific transmembrane protein cadherin, and adherens junction-associated peripheral proteins alpha-catenin, beta-catenin, and p120 catenin is reviewed. P-glycoprotein and some other transporters recently discovered in endothelial cells prevent several compounds from entering the brain parenchyma. It is likely that the transient inhibition of P-glycoprotein by antidepressants enables other medicines to enter the brain. Vesicular transport with clathrin-mediated or adsorptive endocytosis through endothelial cells is also critical for transportation of blood-born substances from the bloodstream to the brain. How medicines pass the BBB to reach the brain parenchyma is discussed.
- Published
- 2007
- Full Text
- View/download PDF
45. Ambient GABA promotes cortical entry of tangentially migrating cells derived from the medial ganglionic eminence.
- Author
-
Cuzon VC, Yeh PW, Cheng Q, and Yeh HH
- Subjects
- Animals, Cells, Cultured, Dose-Response Relationship, Drug, Median Eminence cytology, Median Eminence drug effects, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neocortex cytology, Neocortex drug effects, Neurons drug effects, gamma-Aminobutyric Acid metabolism, Cell Movement drug effects, Median Eminence embryology, Median Eminence physiology, Neocortex embryology, Neocortex physiology, Neurons physiology, gamma-Aminobutyric Acid administration & dosage
- Abstract
During corticogenesis, cells from the medial ganglionic eminence (MGE) migrate tangentially into the neocortical anlage. Here we report that gamma-aminobutyric acid (GABA), via GABAA receptors, regulates tangential migration. In embryonic telencephalic slices, bicuculline produced an outward current in migrating MGE-derived cells in the neocortex, suggesting the presence of and tonic activation by ambient GABA. Ambient GABA was also present in the MGE, although this required demonstration using as bioassay HEK293 cells expressing high-affinity alpha6/beta2/gamma2s recombinant GABAA receptors. The concentration of ambient GABA was 0.5+/-0.1 microM in both regions. MGE-derived cells before the corticostriate juncture (CSJ) were less responsive to GABA than those in the neocortex, and profiling of GABAA receptor subunit transcripts revealed different expression patterns in the MGE vis-à-vis the neocortex. These findings suggest a dynamic expression of GABAA receptor number or isoform as MGE-derived cells enter the neocortex and become tonically influenced by ambient GABA. Treatment with bicuculline or antibody against GABA did not affect migration of MGE-derived cells before the CSJ but decreased "crossing index," reflecting impeded migration past the CSJ into the neocortex. Treatment with diazepam or addition of exogenous GABA increased crossing index. We conclude that ambient GABA promotes cortical entry of tangentially migrating MGE-derived cells.
- Published
- 2006
- Full Text
- View/download PDF
46. Sexual dimorphism in the organization of the rat hypothalamic infundibular area.
- Author
-
Ciofi P, Leroy D, and Tramu G
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus physiology, Female, Immunohistochemistry methods, Male, Median Eminence physiology, Microscopy, Immunoelectron methods, Nerve Tissue Proteins metabolism, Pituitary Hormones metabolism, Rats, Sex Factors, Arcuate Nucleus of Hypothalamus anatomy & histology, Median Eminence anatomy & histology, Sex Characteristics
- Abstract
The hypothalamic infundibular area is located outside the blood-brain barrier and includes, the ventromedial arcuate nucleus (vmARC) sensing circulating substances, and the median eminence (ME) where neurohormones are released into the hypothalamo-hypophysial vasculature. This integrated functional unit, pivotal in endocrine control, adjusts neuroendocrine output to feedback information. Despite a differing physiology in males and females, this functional unit has not appeared differently organized between sexes. Using immunocytochemistry, we describe here for the first time in adult rats, a conspicuous sex-difference in its axonal wiring by intrinsic glutamatergic neurons containing the neuropeptides neurokinin B (NKB) and dynorphin. In the male, NKB neurons send axons to capillary vessels of the vmARC and of the ME (only where gonadotropin-releasing hormone (GnRH) axons terminate). Electron microscopy revealed that NKB axons target the barrier of tanycytes around fenestrated capillary vessels (in addition to GnRH axons), suggesting a control of regional bidirectional permeability. In the female, NKB neurons send axons to the neuropile of the vmARC, suggesting a direct control of its sensor neurons. The other projections of NKB neurons, studied by surgical isolation of the ARC-ME complex and confocal microscopy, are not sexually dimorphic and target both integrative and neuroendocrine centers controlling reproduction and metabolism, suggesting a broad influence over endocrine function. These observations demonstrate that the mechanisms subserving hypothalamic permeability and sensitivity to feedback information are sexually dimorphic, making the infundibular area a privileged site of generation of the male-to-female differences in the adult pattern of pulsatile hormonal secretions.
- Published
- 2006
- Full Text
- View/download PDF
47. Brain-derived neurotrophic factor in the brain of Xenopus laevis may act as a pituitary neurohormone together with mesotocin.
- Author
-
Calle M, Wang L, Kuijpers FJ, Cruijsen PM, Arckens L, and Roubos EW
- Subjects
- Animals, Immunohistochemistry, Median Eminence anatomy & histology, Median Eminence physiology, Melanocytes metabolism, Melanocytes ultrastructure, Microscopy, Immunoelectron, Oxytocin metabolism, Oxytocin pharmacology, Pituitary Gland cytology, Pituitary Gland drug effects, Presynaptic Terminals metabolism, Presynaptic Terminals ultrastructure, alpha-MSH metabolism, Brain-Derived Neurotrophic Factor metabolism, Neurotransmitter Agents metabolism, Oxytocin analogs & derivatives, Pituitary Gland physiology, Xenopus laevis physiology
- Abstract
Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, occurs abundantly in the brain, where it exerts a variety of neural functions. We previously demonstrated that BDNF also exists in the endocrine melanotroph cells in the intermediate lobe of the pituitary gland of the amphibian Xenopus laevis, suggesting that BDNF, in addition to its neural actions within the brain, can act as a hormone. In the present study, we tested whether BDNF, in addition to its neural and hormonal roles, can be released as a neurohormone from the neural pituitary lobe of X. laevis. By light immunocytochemistry, we show that BDNF is present in perikarya, in ventrolaterally projecting axons of the hypothalamic magnocellular nucleus and in the neural lobe of the pituitary gland, and that it coexists in these structures with the amphibian neurohormone, mesotocin. The neural lobe was studied in detail at the ultrastructural level. Two types of neurohaemal axon terminals were observed, occurring intermingled and in similar numbers. Type A is filled with round, moderately electron-dense secretory granules with a mean diameter of approximately 145 nm. Type B terminals contain electron-dense and smaller, ellipsoid granules (long and short diameter approximately 140 and 100 nm, respectively). BDNF is exclusively present in secretory granules of type A axon terminals. Double gold-immunolabelling revealed that BDNF coexists in these granules with mesotocin. Furthermore, we demonstrate in an superfusion study performed in vitro that mesotocin stimulates peptide release from the endocrine melanotroph cells. On the basis of these data, we propose that BDNF can act on these cells as a neurohormone.
- Published
- 2006
- Full Text
- View/download PDF
48. Circadian rhythms of prolactin secretion in neonatal female rabbits after acute separation from their mothers.
- Author
-
Alvarez MP, Jiménez V, Cano P, Rebollar P, Cardinali DP, and Esquifino AI
- Subjects
- Animals, Aspartic Acid metabolism, Dopamine metabolism, Female, Glutamic Acid metabolism, Lactation, Median Eminence physiology, Pituitary Gland, Anterior physiology, Serotonin metabolism, Taurine metabolism, gamma-Aminobutyric Acid metabolism, Animals, Newborn, Circadian Rhythm, Maternal Deprivation, Prolactin metabolism, Rabbits physiology
- Abstract
Newborn rabbits (Oryctolagus cuniculus) are only nursed for 3-5 min every 24 h and show a circadian increase in activity in anticipation of nursing. The objective of this study was to determine, in neonatal female rabbits after acute separation from the doe for 48 h, the changes in 24-h rhythms of plasma prolactin and median eminence and anterior pituitary concentration of dopamine (DA) and serotonin (5HT). In addition, median eminence concentration of the excitatory amino acid transmitters glutamate (GLU) and aspartate (ASP) and of the inhibitory amino acid transmitters gamma-aminobutyric acid (GABA) and taurine (TAU) was measured. A significant 21% increase of circulating prolactin occurred in isolated pups. In controls pups, plasma prolactin levels showed two peaks, during the first half of the light phase and at the beginning of the scotophase, respectively. In the isolated pups, a phase advance of about 4 h occurred for the two prolactin peaks. Hemicircadian changes of median eminence DA were found in controls, whereas a single daily peak (at 17:00 h) was found in the separated pups. Plasma prolactin and median eminence DA correlated significantly and inversely in the control group only. Pituitary DA content exhibited a single peak in controls and a hemicircadian pattern in isolated pups. Plasma prolactin and pituitary DA correlated significantly in isolated pups only 00000. Pup isolation decreased median eminence 5HT levels, augmented pituitary 5HT levels and disrupted their 24 h rhythmicity. Circulating prolactin correlated inversely with median eminence 5HT and directly with adenohypophysial 5HT only in controls. Isolation of pups generally modified the 24 h pattern of median eminence excitatory and inhibitory amino acid content by causing a prominent decrease at the beginning of the light phase. The results indicate that circadian rhythmicity of prolactin secretory mechanisms in female rabbit pups is significantly affected by pup's isolation from the doe.
- Published
- 2006
- Full Text
- View/download PDF
49. Expression of a dominant negative FGF receptor in developing GNRH1 neurons disrupts axon outgrowth and targeting to the median eminence.
- Author
-
Gill JC and Tsai PS
- Subjects
- Animals, Cells, Cultured, Coculture Techniques, Female, Fibroblast Growth Factor 2 metabolism, Fibroblast Growth Factor 2 pharmacology, Green Fluorescent Proteins genetics, Male, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Transgenic, Microspheres, Neural Pathways physiology, Neurons physiology, Neurons ultrastructure, Pregnancy, Signal Transduction physiology, Stilbamidines, Axons physiology, Gonadotropin-Releasing Hormone metabolism, Median Eminence cytology, Median Eminence embryology, Median Eminence physiology, Protein Precursors metabolism, Receptor, Fibroblast Growth Factor, Type 1 genetics, Receptor, Fibroblast Growth Factor, Type 1 metabolism
- Abstract
During development, neurons that synthesize and release gonadotropin-releasing hormone (GNRH1) extend their axons to the median eminence (ME) to establish neurosecretory contacts necessary for hormone secretion. Signals that coordinate this process are not known, but could involve the activation of fibroblast growth factor receptors (FGFRs) expressed on developing GNRH1 neurons. Using both whole-animal and cell culture approaches, this study examines the direct role of FGFR signaling in the extension and guidance of GNRH1 axons to the ME. In vivo retrograde labeling with fluorogold (FG) first showed a significant reduction in the projections of GNRH1 axons to the circumventricular organs (including the ME) in transgenic mice expressing a dominant negative FGF receptor (dnFGFR) in GNRH1 neurons. Using a primary GNRH1 neuronal culture system, we examined if compromised axon extension and directional growth led to the reduced axon targeting efficiency seen in vivo. Primary cultures of GNRH1 neurons were established from Embryonic Day 15.5 embryos, an age when GNRH1 neurons are actively targeting the ME. Cultured GNRH1 neurons expressing dnFGFR (dnFGFR neurons) exhibited attenuated activation of signaling pathways and reduced neurite outgrowth in response to FGF2. Further, dnFGFR neurons failed to preferentially target neurites toward cocultured ME explant and FGF2-coated beads, suggesting a defect in axon pathfinding. Together, these findings describe a direct role of FGFR signaling in the elongation and guidance of GNRH1 axons to the ME.
- Published
- 2006
- Full Text
- View/download PDF
50. Ventromedial arcuate nucleus communicates peripheral metabolic information to the suprachiasmatic nucleus.
- Author
-
Yi CX, van der Vliet J, Dai J, Yin G, Ru L, and Buijs RM
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus cytology, Cell Communication physiology, Cholera Toxin, Male, Median Eminence physiology, Microscopy, Confocal, Neurons cytology, Rats, Rats, Wistar, Suprachiasmatic Nucleus cytology, Arcuate Nucleus of Hypothalamus physiology, Neurons physiology, Suprachiasmatic Nucleus physiology
- Abstract
The arcuate nucleus (ARC) is crucial for the maintenance of energy homeostasis as an integrator of long- and short-term hunger and satiety signals. The expression of receptors for metabolic hormones, such as insulin, leptin, and ghrelin, allows ARC to sense information from the periphery and signal it to the central nervous system. The ventromedial ARC (vmARC) mainly comprises orexigenic neuropeptide agouti-related peptide and neuropeptide Y neurons, which are sensitive to circulating signals. To investigate neural connections of vmARC within the central nervous system, we injected the neuronal tracer cholera toxin B into vmARC. Due to variation of injection sites, tracer was also injected into the subependymal layer of the median eminence (seME), which showed similar projection patterns as the vmARC. We propose that the vmARC forms a complex with the seME, their reciprocal connections with viscerosensory areas in brain stem, and other circumventricular organs, suggesting the exchange of metabolic and circulating information. For the first time, the vmARC-seME was shown to have reciprocal interaction with the suprachiasmatic nucleus (SCN). Activation of vmARC neurons by systemic administration of the ghrelin mimetic GH-releasing peptide-6 combined with SCN tracing showed vmARC neurons to transmit feeding related signals to the SCN. The functionality of this pathway was demonstrated by systemic injection of GH-releasing peptide-6, which induced Fos in the vmARC and resulted in a reduction of about 40% of early daytime Fos immunoreactivity in the SCN. This observation suggests an anatomical and functional pathway for peripheral hormonal feedback to the hypothalamus, which may serve to modulate the activity of the SCN.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.