1
|
Wu J, Yang OJ, Soderblom EJ, Yan D. Heat Shock Proteins Function as Signaling Molecules to Mediate Neuron-Glia Communication During Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576052. [PMID: 38293019 PMCID: PMC10827141 DOI: 10.1101/2024.01.18.576052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The nervous system is primarily composed of neurons and glia, and the communication between them plays profound roles in regulating the development and function of the brain. Neuron-glia signal transduction is known to be mediated by secreted or juxtacrine signals through ligand-receptor interactions on the cell membrane. Here, we report a novel mechanism for neuron-glia signal transduction, wherein neurons transmit proteins to glia through extracellular vesicles, activating glial signaling pathways. We find that in the amphid sensory organ of Caenorhabditis elegans, different sensory neurons exhibit varying aging rates. This discrepancy in aging is governed by the crosstalk between neurons and glia. We demonstrate that early-aged neurons can transmit heat shock proteins (HSP) to glia via extracellular vesicles. These neuronal HSPs activate the IRE1-XBP1 pathway, further increasing their expression in glia, forming a positive feedback loop. Ultimately, the activation of the IRE1-XBP-1 pathway leads to the transcriptional regulation of chondroitin synthases to protect glia-embedded neurons from aging-associated functional decline. Therefore, our studies unveil a novel mechanism for neuron-glia communication in the nervous system and provide new insights into our understanding of brain aging.
Collapse
Affiliation(s)
- Jieyu Wu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Olivia Jiaming Yang
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
- East Chapel Hill High School, Chapel Hill, NC 27514, USA
| | - Erik J. Soderblom
- Proteomics and Metabolomics Core Facility, Duke University Medical School, Durham, NC 27710, USA
| | - Dong Yan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell biology, Department of Neurobiology, Regeneration next, and Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
2
|
Prevot V, Sharif A. The polygamous GnRH neuron: Astrocytic and tanycytic communication with a neuroendocrine neuronal population. J Neuroendocrinol 2022; 34:e13104. [PMID: 35233849 DOI: 10.1111/jne.13104] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/12/2022] [Accepted: 01/30/2022] [Indexed: 11/28/2022]
Abstract
To ensure the survival of the species, hypothalamic neuroendocrine circuits controlling fertility, which converge onto neurons producing gonadotropin-releasing hormone (GnRH), must respond to fluctuating physiological conditions by undergoing rapid and reversible structural and functional changes. However, GnRH neurons do not act alone, but through reciprocal interactions with multiple hypothalamic cell populations, including several glial and endothelial cell types. For instance, it has long been known that in the hypothalamic median eminence, where GnRH axons terminate and release their neurohormone into the pituitary portal blood circulation, morphological plasticity displayed by distal processes of tanycytes modifies their relationship with adjacent neurons as well as the spatial properties of the neurohemal junction. These alterations not only regulate the capacity of GnRH neurons to release their neurohormone, but also the activation of discrete non-neuronal pathways that mediate feedback by peripheral hormones onto the hypothalamus. Additionally, a recent breakthrough has demonstrated that GnRH neurons themselves orchestrate the establishment of their neuroendocrine circuitry during postnatal development by recruiting an entourage of newborn astrocytes that escort them into adulthood and, via signalling through gliotransmitters such as prostaglandin E2, modulate their activity and GnRH release. Intriguingly, several environmental and behavioural toxins perturb these neuron-glia interactions and consequently, reproductive maturation and fertility. Deciphering the communication between GnRH neurons and other neural cell types constituting hypothalamic neuroendocrine circuits is thus critical both to understanding physiological processes such as puberty, oestrous cyclicity and aging, and to developing novel therapeutic strategies for dysfunctions of these processes, including the effects of endocrine disruptors.
Collapse
Affiliation(s)
- Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| |
Collapse
|
3
|
GnRH neurons recruit astrocytes in infancy to facilitate network integration and sexual maturation. Nat Neurosci 2021; 24:1660-1672. [PMID: 34795451 DOI: 10.1038/s41593-021-00960-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 10/08/2021] [Indexed: 12/25/2022]
Abstract
Neurons that produce gonadotropin-releasing hormone (GnRH), which control fertility, complete their nose-to-brain migration by birth. However, their function depends on integration within a complex neuroglial network during postnatal development. Here, we show that rodent GnRH neurons use a prostaglandin D2 receptor DP1 signaling mechanism during infancy to recruit newborn astrocytes that 'escort' them into adulthood, and that the impairment of postnatal hypothalamic gliogenesis markedly alters sexual maturation by preventing this recruitment, a process mimicked by the endocrine disruptor bisphenol A. Inhibition of DP1 signaling in the infantile preoptic region, where GnRH cell bodies reside, disrupts the correct wiring and firing of GnRH neurons, alters minipuberty or the first activation of the hypothalamic-pituitary-gonadal axis during infancy, and delays the timely acquisition of reproductive capacity. These findings uncover a previously unknown neuron-to-neural-progenitor communication pathway and demonstrate that postnatal astrogenesis is a basic component of a complex set of mechanisms used by the neuroendocrine brain to control sexual maturation.
Collapse
|
4
|
Maione L, Bouvattier C, Kaiser UB. Central precocious puberty: Recent advances in understanding the aetiology and in the clinical approach. Clin Endocrinol (Oxf) 2021; 95:542-555. [PMID: 33797780 PMCID: PMC8586890 DOI: 10.1111/cen.14475] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
Central precocious puberty (CPP) results from early activation of the hypothalamic-pituitary-gonadal (HPG) axis. The current state of knowledge of the complex neural network acting at the level of the hypothalamus and the GnRH neuron to control puberty onset has expanded, particularly in the context of molecular interactions. Along with these advances, the knowledge of pubertal physiology and pathophysiology has also increased. This review focuses on regulatory abnormalities occurring at the hypothalamic level of the HPG axis to cause CPP. The clinical approach to diagnosis of puberty and pubertal disorders is also reviewed, with a particular focus on aetiologies of CPP. The recent identification of mutations in MKRN3 and DLK1 in familial as well sporadic forms of CPP has changed the state of the art of the approach to patients with CPP. Genetic advances have also had important repercussions beyond consideration of puberty alone. Syndromic disorders and central nervous system lesions associated with CPP are also discussed. If untreated, these conditions may lead to adverse physical, psychosocial and medical outcomes.
Collapse
Affiliation(s)
- Luigi Maione
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse, Université Paris-Saclay, Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Claire Bouvattier
- Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse, Université Paris-Saclay, Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Ursula B. Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Identification of Circular RNAs in Hypothalamus of Gilts during the Onset of Puberty. Genes (Basel) 2021; 12:genes12010084. [PMID: 33445426 PMCID: PMC7827264 DOI: 10.3390/genes12010084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 01/09/2023] Open
Abstract
The disorders of puberty have shown negative outcomes on health of mammals, and the hypothalamus is thought to be the main regulator of puberty by releasing GnRH. Many studies show that the circular RNAs (circRNAs) might be implicated in the timing of puberty in mammals. However, the circRNAs in the hypothalamus of gilts have not been explored. To profile the changes and biological functions of circRNAs in the hypothalamus during the onset of puberty, RNA-seq was utilized to establish pre-, in-, and post-pubertal hypothalamic circRNAs profiles. In this study, the functions of hypothalamic circRNAs were enriched in the signaling pathway of neurotrophin, progesterone-mediated oocyte maturation, oocyte meiosis, insulin, ErbB, and mTOR, which have been highly suggested to be involved in the timing of puberty. Furthermore, 53 circRNAs were identified to be putative hypothalamus-specific expressed circRNAs, and some of them were exclusively expressed in the one of three pubertal stages. Moreover, 22 differentially expressed circRNAs were identified and chosen to construct the circRNA-miRNA-gene network. Moreover, 10 circRNAs were found to be driven by six puberty-related genes (ESR1, NF1, APP, ENPP2, ARNT, and DICER1). Subsequently, the expression changes of several circRNAs were confirmed by RT-qPCR. Collectively, the preliminary results of hypothalamic circRNAs provided useful information for the investigation of the molecular mechanism for the timing of puberty in gilts.
Collapse
|
6
|
Smedlund KB, Hill JW. The role of non-neuronal cells in hypogonadotropic hypogonadism. Mol Cell Endocrinol 2020; 518:110996. [PMID: 32860862 DOI: 10.1016/j.mce.2020.110996] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/01/2020] [Accepted: 08/16/2020] [Indexed: 12/18/2022]
Abstract
The hypothalamic-pituitary-gonadal axis is controlled by gonadotropin-releasing hormone (GnRH) released by the hypothalamus. Disruption of this system leads to impaired reproductive maturation and function, a condition known as hypogonadotropic hypogonadism (HH). Most studies to date have focused on genetic causes of HH that impact neuronal development and function. However, variants may also impact the functioning of non-neuronal cells known as glia. Glial cells make up 50% of brain cells of humans, primates, and rodents. They include radial glial cells, microglia, astrocytes, tanycytes, oligodendrocytes, and oligodendrocyte precursor cells. Many of these cells influence the hypothalamic neuroendocrine system controlling fertility. Indeed, glia regulate GnRH neuronal activity and secretion, acting both at their cell bodies and their nerve endings. Recent work has also made clear that these interactions are an essential aspect of how the HPG axis integrates endocrine, metabolic, and environmental signals to control fertility. Recognition of the clinical importance of interactions between glia and the GnRH network may pave the way for the development of new treatment strategies for dysfunctions of puberty and adult fertility.
Collapse
Affiliation(s)
- Kathryn B Smedlund
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
7
|
Qi Z, Mi C, Wu F, Yang X, Sang Y, Liu Y, Li J, Yang H, Xu B, Liu W, Xu Z, Deng Y. The effect of manganese exposure on GnRH secretion via Nrf2/mGluR5/COX-2/PGE2/signaling pathway. Toxicol Ind Health 2020; 35:211-227. [PMID: 30862296 DOI: 10.1177/0748233719825720] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There are limited studies focused on the precise mechanism of gonadotropin-releasing hormone (GnRH) secretion dysfunction after overexposure to manganese (Mn). The objective of the present study was to explore the mechanism of Mn disruption of GnRH synthesis via nuclear factor erythroid-2-related factor-2 (Nrf2)/metabotropic glutamate receptor-5 (mGluR5)/cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) signaling pathway in vitro and in vivo. Primary astrocytes were cultured and treated with different doses of Mn, tert-butylhydroquinonet (tBHQ; Nrf2 agonists), 3-[(2-methyl-4-thaizolyl) ethynyl] pyridine (MTEP; mGluR5 inhibitor), and celecoxib (COX-2 inhibitor) to measure the levels of COX-2, mGluR5, Nrf2, and Nrf2 target genes. Mice were randomly divided into 11 groups, of which included the control group, 12.5-, 25-, and 50-mg/kg MnCl2 group, dimethyl sulfoxide (DMSO) group, tBHQ control group, tBHQ pretreatment group, MTEP control group, MTEP pretreatment group, celecoxib control group, and celecoxib pretreatment group. The injection was administered every day for 2 weeks. Then, levels of GnRH, PGE2, COX-2, mGluR5, Nrf2, Nrf2 target genes, and morphological changes in the hypothalamus of mice were measured. Mn reduced protein levels of Nrf2 and mRNA expression of Nrf2 target genes and increased mGluR5, COX-2, PGE2, and GnRH levels. Meanwhile, injury-related histomorphology changes in the hypothalamus of mice were significantly present. In conclusion, excessive exposure to Mn disrupts GnRH secretion through Nrf2/mGluR5/COX-2/PGE2 signaling pathway.
Collapse
Affiliation(s)
- Zhipeng Qi
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Chao Mi
- 2 Department of School Health Supervision, Institute of Shenyang Health Inspection, Shenyang, People's Republic of China
| | - Fengdi Wu
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Xinxin Yang
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Yanqi Sang
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Yanan Liu
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Jiashuo Li
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Haibo Yang
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China.,3 Department of Occupational Diseases, Linyi People's Hospital, Shandong, People's Republic of China
| | - Bin Xu
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Wei Liu
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Zhaofa Xu
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| | - Yu Deng
- 1 Department of Environmental Health, School of Public Health, China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
8
|
Moeller-Gnangra H, Ernst J, Pfeifer M, Heger S. ErbB4 point mutation in CU3 inbred rats affects gonadotropin-releasing-hormone neuronal function via compromised neuregulin-stimulated prostaglandin E2 release from astrocytes. Glia 2018; 67:309-320. [PMID: 30485552 DOI: 10.1002/glia.23541] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/26/2018] [Accepted: 09/14/2018] [Indexed: 11/05/2022]
Abstract
Gonadotropin releasing hormone (GnRH)-secretion is not only regulated by neuronal factors but also by astroglia cells via growth factors and ErbB receptors of the epidermal growth factor family. Studies in transgenic mice carrying mutations in the ErbB receptor system experience impaired reproductive capacity. In addition, some of these animals show a typical skin phenotype with wavy hair and curly whiskers. The rat strain SPRD-CU3 (CU3), examined in this study, displays a similar skin phenotype and a significant impairment of the timing of puberty onset and reproductive performance, suggesting a disruption in the astrocytic to GnRH neuronal communication. To address this issue, we analyzed astrocytic prostaglandin E2 (PGE2 ) release from primary hypothalamic astrocytic cell cultures after stimulation with transforming growth factor α (TGFα), ligand for ErbB1/ErbB2, or Neuregulin 1 beta 2 (NRG1ß2 ), ligand for ErbB4/ErbB2 signaling pathway. Compared to cultures from wild type animals, astrocytic cultures from CU3 rats were unable to respond to NRG stimulation, suggesting a disruption of the ErbB4/ErbB2 signaling pathway. This is confirmed by mutational analysis of ErbB4 that revealed a single point mutation at 3125 bp resulting in an amino acid change from proline to glutamine located at the carboxy-terminal region. As a consequence, substantial conformational changes occur in the transmembrane and intracellular domain of the protein, affecting the ability to form a receptor dimer with a partner and the ability to function as a transcriptional regulator. Thus, astroglia to GnRH neuronal signaling via ErbB4 is essential of timely onset of puberty and reproductive function.
Collapse
Affiliation(s)
| | - Johanna Ernst
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Manuel Pfeifer
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Sabine Heger
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany.,Children's Hospital "Auf der Bult", Department of Pediatrics, Hannover, Germany
| |
Collapse
|
9
|
Prevot V, Dehouck B, Sharif A, Ciofi P, Giacobini P, Clasadonte J. The Versatile Tanycyte: A Hypothalamic Integrator of Reproduction and Energy Metabolism. Endocr Rev 2018; 39:333-368. [PMID: 29351662 DOI: 10.1210/er.2017-00235] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
The fertility and survival of an individual rely on the ability of the periphery to promptly, effectively, and reproducibly communicate with brain neural networks that control reproduction, food intake, and energy homeostasis. Tanycytes, a specialized glial cell type lining the wall of the third ventricle in the median eminence of the hypothalamus, appear to act as the linchpin of these processes by dynamically controlling the secretion of neuropeptides into the portal vasculature by hypothalamic neurons and regulating blood-brain and blood-cerebrospinal fluid exchanges, both processes that depend on the ability of these cells to adapt their morphology to the physiological state of the individual. In addition to their barrier properties, tanycytes possess the ability to sense blood glucose levels, and play a fundamental and active role in shuttling circulating metabolic signals to hypothalamic neurons that control food intake. Moreover, accumulating data suggest that, in keeping with their putative descent from radial glial cells, tanycytes are endowed with neural stem cell properties and may respond to dietary or reproductive cues by modulating hypothalamic neurogenesis. Tanycytes could thus constitute the missing link in the loop connecting behavior, hormonal changes, signal transduction, central neuronal activation and, finally, behavior again. In this article, we will examine these recent advances in the understanding of tanycytic plasticity and function in the hypothalamus and the underlying molecular mechanisms. We will also discuss the putative involvement and therapeutic potential of hypothalamic tanycytes in metabolic and fertility disorders.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Bénédicte Dehouck
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Ariane Sharif
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Philippe Ciofi
- Inserm, Neurocentre Magendie, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| |
Collapse
|
10
|
Clasadonte J, Prevot V. The special relationship: glia-neuron interactions in the neuroendocrine hypothalamus. Nat Rev Endocrinol 2018; 14:25-44. [PMID: 29076504 DOI: 10.1038/nrendo.2017.124] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Natural fluctuations in physiological conditions require adaptive responses involving rapid and reversible structural and functional changes in the hypothalamic neuroendocrine circuits that control homeostasis. Here, we discuss the data that implicate hypothalamic glia in the control of hypothalamic neuroendocrine circuits, specifically neuron-glia interactions in the regulation of neurosecretion as well as neuronal excitability. Mechanistically, the morphological plasticity displayed by distal processes of astrocytes, pituicytes and tanycytes modifies the geometry and diffusion properties of the extracellular space. These changes alter the relationship between glial cells of the hypothalamus and adjacent neuronal elements, especially at specialized intersections such as synapses and neurohaemal junctions. The structural alterations in turn lead to functional plasticity that alters the release and spread of neurotransmitters, neuromodulators and gliotransmitters, as well as the activity of discrete glial signalling pathways that mediate feedback by peripheral signals to the hypothalamus. An understanding of the contributions of these and other non-neuronal cell types to hypothalamic neuroendocrine function is thus critical both to understand physiological processes such as puberty, the maintenance of bodily homeostasis and ageing and to develop novel therapeutic strategies for dysfunctions of these processes, such as infertility and metabolic disorders.
Collapse
Affiliation(s)
- Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Bâtiment Biserte, 1 Place de Verdun, 59045, Lille, Cedex, France
- University of Lille, FHU 1000 days for Health, School of Medicine, Lille 59000, France
| | - Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Bâtiment Biserte, 1 Place de Verdun, 59045, Lille, Cedex, France
- University of Lille, FHU 1000 days for Health, School of Medicine, Lille 59000, France
| |
Collapse
|
11
|
Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol 2016; 144:5-26. [DOI: 10.1016/j.pneurobio.2016.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
|
12
|
Abstract
The gonadotropin-releasing hormone (GnRH) neuronal network generates pulse and surge modes of gonadotropin secretion critical for puberty and fertility. The arcuate nucleus kisspeptin neurons that innervate the projections of GnRH neurons in and around their neurosecretory zone are key components of the pulse generator in all mammals. By contrast, kisspeptin neurons located in the preoptic area project to GnRH neuron cell bodies and proximal dendrites and are involved in surge generation in female rodents (and possibly other species). The hypothalamic-pituitary-gonadal axis develops embryonically but, apart from short periods of activation immediately after birth, remains suppressed through a combination of gonadal and non-gonadal mechanisms. At puberty onset, the pulse generator reactivates, probably owing to progressive stimulatory influences on GnRH neurons from glial and neurotransmitter signalling, and the re-emergence of stimulatory arcuate kisspeptin input. In females, the development of pulsatile gonadotropin secretion enables final maturation of the surge generator that ultimately triggers the first ovulation. Representation of the GnRH neuronal network as a series of interlocking functional modules could help conceptualization of its functioning in different species. Insights into pulse and surge generation are expected to aid development of therapeutic strategies ameliorating pubertal disorders and infertility in the clinic.
Collapse
Affiliation(s)
- Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| |
Collapse
|
13
|
Panatier A, Robitaille R. Astrocytic mGluR5 and the tripartite synapse. Neuroscience 2015; 323:29-34. [PMID: 25847307 DOI: 10.1016/j.neuroscience.2015.03.063] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 03/19/2015] [Accepted: 03/26/2015] [Indexed: 11/27/2022]
Abstract
In the brain, astrocytes occupy a key position between vessels and synapses. Among their numerous functions, these glial cells are key partners of neurons during synaptic transmission. Astrocytes detect transmitter release through receptors and transporters at the level of their processes, which are in close proximity to the tow neuronal elements of synapses. In response to transmitter-mediated activation, glial cells in turn regulate synaptic transmission and neuronal excitability. This process has been reported to involve several glial receptors. One of the best known of such receptors is the metabotropic glutamatergic receptor subtype 5 (mGluR5). In the present review we will discuss the implication of mGluR5s as detectors of synaptic transmission. In particular, we will discuss how the functional properties and localization of these receptors permit the detection of the synaptic signal in a defined temporal window and a given spatial area around the synapse. Furthermore, we will review the impact of their activation on synaptic transmission.
Collapse
Affiliation(s)
- A Panatier
- Neurocentre Magendie, INSERM U862, Bordeaux, France; Université de Bordeaux, Bordeaux, France.
| | - R Robitaille
- Groupe de recherche sur le système nerveux central, Université de Montréal, Canada; Département de neurosciences, Université de Montréal, PO Box 6128, Station centre-ville, Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
14
|
Goudriaan A, de Leeuw C, Ripke S, Hultman CM, Sklar P, Sullivan PF, Smit AB, Posthuma D, Verheijen MHG. Specific glial functions contribute to schizophrenia susceptibility. Schizophr Bull 2014; 40:925-35. [PMID: 23956119 PMCID: PMC4059439 DOI: 10.1093/schbul/sbt109] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a highly polygenic brain disorder. The main hypothesis for disease etiology in schizophrenia primarily focuses on the role of dysfunctional synaptic transmission. Previous studies have therefore directed their investigations toward the role of neuronal dysfunction. However, recent studies have shown that apart from neurons, glial cells also play a major role in synaptic transmission. Therefore, we investigated the potential causal involvement of the 3 principle glial cell lineages in risk to schizophrenia. We performed a functional gene set analysis to test for the combined effects of genetic variants in glial type-specific genes for association with schizophrenia. We used genome-wide association data from the largest schizophrenia sample to date, including 13 689 cases and 18 226 healthy controls. Our results show that astrocyte and oligodendrocyte gene sets, but not microglia gene sets, are associated with an increased risk for schizophrenia. The astrocyte and oligodendrocyte findings are related to astrocyte signaling at the synapse, myelin membrane integrity, glial development, and epigenetic control. Together, these results show that genetic alterations underlying specific glial cell type functions increase susceptibility to schizophrenia and provide evidence that the neuronal hypothesis of schizophrenia should be extended to include the role of glia.
Collapse
Affiliation(s)
- Andrea Goudriaan
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, the Netherlands;
| | - Christiaan de Leeuw
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, the Netherlands; Institute for Computing and Information Sciences, Radboud University, Nijmegen, the Netherlands
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| | - Christina M Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Pamela Sklar
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Center for Psychiatric Genomics, Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, the Netherlands
| | - Danielle Posthuma
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, the Netherlands;
| | - Mark H G Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, the Netherlands
| |
Collapse
|
15
|
Focant MC, Goursaud S, Boucherie C, Dumont AO, Hermans E. PICK1 expression in reactive astrocytes within the spinal cord of amyotrophic lateral sclerosis (ALS) rats. Neuropathol Appl Neurobiol 2013; 39:231-42. [PMID: 22624977 DOI: 10.1111/j.1365-2990.2012.01282.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIMS The protein interacting with C kinase 1 (PICK1), a PDZ domain-containing protein mainly expressed in the central nervous system, interacts with the glutamate receptor subunit GluR2, with the glutamate transporter GLT-1b and with the enzyme serine racemase. These three proteins appear as key actors in the glutamate-mediated excitotoxicity associated with amyotrophic lateral sclerosis (ALS), in both patients and animal models of the disease. In this study, we examined the expression of PICK1 in the spinal cord of transgenic rats expressing a mutated form of the human superoxide dismutase 1 (hSOD1(G93A) ) during the progression of the disease. METHODS Expression of PICK1 was examined by real-time qPCR at presymptomatic and symptomatic stages as well as at end-stage. The expression of PICK1 in the different cell types of the spinal cord was examined by immunohistochemistry. RESULTS The overall expression of PICK1 is not modified in cervical and lumbar spinal cord of transgenic (hSOD1(G93A) ) rats during the progression of the disease. Nonetheless, immunohistochemical studies of lumbar ventral horns revealed a shift of PICK1 expression from motor neurones in healthy rats to activated astrocytes in end-stage hSOD1(G93A) animals. CONCLUSIONS Considering the documented influence of PICK1 expression on d-serine release and glutamate transport in astrocytes, these findings point to a potential implication of PICK1 in the progression of ALS.
Collapse
Affiliation(s)
- M C Focant
- Group of Neuropharmacology, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | | | | | | | | |
Collapse
|
16
|
Bradley SJ, Challiss RJ. G protein-coupled receptor signalling in astrocytes in health and disease: A focus on metabotropic glutamate receptors. Biochem Pharmacol 2012; 84:249-59. [DOI: 10.1016/j.bcp.2012.04.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 04/02/2012] [Accepted: 04/09/2012] [Indexed: 02/03/2023]
|
17
|
Sandau US, Alderman Z, Corfas G, Ojeda SR, Raber J. Astrocyte-specific disruption of SynCAM1 signaling results in ADHD-like behavioral manifestations. PLoS One 2012; 7:e36424. [PMID: 22558465 PMCID: PMC3340339 DOI: 10.1371/journal.pone.0036424] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 04/02/2012] [Indexed: 11/18/2022] Open
Abstract
SynCAM1 is an adhesion molecule involved in synaptic differentiation and organization. SynCAM1 is also expressed in astroglial cells where it mediates astrocyte-to astrocyte and glial-neuronal adhesive communication. In astrocytes, SynCAM1 is functionally linked to erbB4 receptors, which are involved in the control of both neuronal/glial development and mature neuronal and glial function. Here we report that mice carrying a dominant-negative form of SynCAM1 specifically targeted to astrocytes (termed GFAP-DNSynCAM1 mice) exhibit disrupted diurnal locomotor activity with enhanced and more frequent episodes of activity than control littermates during the day (when the animals are normally sleeping) accompanied by shorter periods of rest. GFAP-DNSynCAM1 mice also display high levels of basal activity in the dark period (the rodent's awake/active time) that are attenuated by the psychostimulant D,L-amphetamine, and reduced anxiety levels in response to both avoidable and unavoidable provoking stimuli. These results indicate that disruption of SynCAM1-dependent astroglial function results in behavioral abnormalities similar to those described in animals model of attention-deficit hyperactive disorder (ADHD), and suggest a hitherto unappreciated contribution of glial cells to the pathophysiology of this disorder.
Collapse
Affiliation(s)
- Ursula S. Sandau
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Zefora Alderman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Gabriel Corfas
- F. M. Kirby Neurobiology Program, Harvard Medical School, Children's Hospital, Boston, Massachusetts, United States of America
| | - Sergio R. Ojeda
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
- * E-mail: (SRO); (JR)
| | - Jacob Raber
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Departments of Behavioral Neurosciences and Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail: (SRO); (JR)
| |
Collapse
|
18
|
Glanowska KM, Moenter SM. Endocannabinoids and prostaglandins both contribute to GnRH neuron-GABAergic afferent local feedback circuits. J Neurophysiol 2011; 106:3073-81. [PMID: 21917995 DOI: 10.1152/jn.00046.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons form the final common pathway for central control of fertility. Regulation of GnRH neurons by long-loop gonadal steroid feedback through steroid receptor-expressing afferents such as GABAergic neurons is well studied. Recently, local central feedback circuits regulating GnRH neurons were identified. GnRH neuronal depolarization induces short-term inhibition of their GABAergic afferents via a mechanism dependent on metabotropic glutamate receptor (mGluR) activation. GnRH neurons are enveloped in astrocytes, which express mGluRs. GnRH neurons also produce endocannabinoids, which can be induced by mGluR activation. We hypothesized the local GnRH-GABA circuit utilizes glia-derived and/or cannabinoid mechanisms and is altered by steroid milieu. Whole cell voltage-clamp was used to record GABAergic postsynaptic currents (PSCs) from GnRH neurons before and after action potential-like depolarizations were mimicked. In GnRH neurons from ovariectomized (OVX) mice, this depolarization reduced PSC frequency. This suppression was blocked by inhibition of prostaglandin synthesis with indomethacin, by a prostaglandin receptor antagonist, or by a specific glial metabolic poison, together suggesting the postulate that prostaglandins, potentially glia-derived, play a role in this circuit. This circuit was also inhibited by a CB1 receptor antagonist or by blockade of endocannabinoid synthesis in GnRH neurons, suggesting an endocannabinoid element, as well. In females, local circuit inhibition persisted in androgen-treated mice but not in estradiol-treated mice or young ovary-intact mice. In contrast, local circuit inhibition was present in gonad-intact males. These data suggest GnRH neurons interact with their afferent neurons using multiple mechanisms and that these local circuits can be modified by both sex and steroid feedback.
Collapse
Affiliation(s)
- Katarzyna M Glanowska
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | | |
Collapse
|
19
|
Prostaglandin E2 release from astrocytes triggers gonadotropin-releasing hormone (GnRH) neuron firing via EP2 receptor activation. Proc Natl Acad Sci U S A 2011; 108:16104-9. [PMID: 21896757 DOI: 10.1073/pnas.1107533108] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Astrocytes in the hypothalamus release prostaglandin E(2) (PGE(2)) in response to cell-cell signaling initiated by neurons and glial cells. Upon release, PGE(2) stimulates the secretion of gonadotropin-releasing hormone (GnRH), the neuropeptide that controls reproduction, from hypothalamic neuroendocrine neurons. Whether this effect on GnRH secretion is accompanied by changes in the firing behavior of these neurons is unknown. Using patch-clamp recording we demonstrate that PGE(2) exerts a dose-dependent postsynaptic excitatory effect on GnRH neurons. These effects are mimicked by an EP2 receptor agonist and attenuated by protein kinase A (PKA) inhibitors. The acute blockade of prostaglandin synthesis by indomethacin (INDO) or the selective inhibition of astrocyte metabolism by fluoroacetate (FA) suppresses the spontaneous firing activity of GnRH neurons in brain slices. Similarly, GnRH neuronal activity is reduced in mice with impaired astrocytic PGE(2) release due to defective erbB signaling in astrocytes. These results indicate that astrocyte-to-neuron communication in the hypothalamus is essential for the activity of GnRH neurons and suggest that PGE(2) acts as a gliotransmitter within the GnRH neurosecretory system.
Collapse
|
20
|
Sandau US, Mungenast AE, Alderman Z, Sardi SP, Fogel AI, Taylor B, Parent AS, Biederer T, Corfas G, Ojeda SR. SynCAM1, a synaptic adhesion molecule, is expressed in astrocytes and contributes to erbB4 receptor-mediated control of female sexual development. Endocrinology 2011; 152:2364-76. [PMID: 21486934 PMCID: PMC3100629 DOI: 10.1210/en.2010-1435] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Female sexual maturation requires erythroblastosis B (erbB)4 signaling in hypothalamic astrocytes; however, the mechanisms by which erbB4 contributes to this process are incompletely understood. Here we show that SynCAM1, a synaptic adhesion molecule with signaling capabilities, is not only expressed highly in neurons, but also in hypothalamic astrocytes and is functionally associated with erbB4 receptor activity. Whereas SynCAM1 expression is diminished in astrocytes with impaired erbB4 signaling, ligand-dependent activation of astroglial erbB4 receptors results in rapid association of erbB4 with SynCAM1 and activation of SynCAM1 gene transcription. To determine whether astrocytic SynCAM1-dependent intracellular signaling is required for normal female reproductive function, we generated transgenic mice that express in an astrocyte-specific manner a dominant-negative form of SynCAM1 lacking the intracellular domain. The mutant protein was correctly targeted to the cell membrane and was functionally viable as shown by its ability to block intracellular calcium/calmodulin-dependent serine protein kinase redistribution, a major SynCAM1-mediated event. Dominant-negative-SynCAM1 female mice had a delayed onset of puberty, disrupted estrous cyclicity, and reduced fecundity. These deficits were associated with a reduced capacity of neuregulin-dependent erbB4 receptor activation to elicit prostaglandin E2 release from astrocytes and GnRH release from the hypothalamus. We conclude that one of the mechanisms underlying erbB4 receptor-mediated facilitation of glial-neuronal interactions in the neuroendocrine brain involves SynCAM1-dependent signaling and that this interaction is required for normal female reproductive function.
Collapse
Affiliation(s)
- Ursula S Sandau
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lenz KM, Wright CL, Martin RC, McCarthy MM. Prostaglandin E₂ regulates AMPA receptor phosphorylation and promotes membrane insertion in preoptic area neurons and glia during sexual differentiation. PLoS One 2011; 6:e18500. [PMID: 21490976 PMCID: PMC3072395 DOI: 10.1371/journal.pone.0018500] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 03/02/2011] [Indexed: 11/19/2022] Open
Abstract
Sexual differentiation of the rodent brain is dependent upon the organizing actions of the steroid hormone, estradiol. In the preoptic area, a brain region critical for the expression of adult reproductive behavior, there are twice as many dendritic spine synapses per unit length on newborn male neurons compared to female neurons and this sex difference correlates with the expression of adult male copulatory behavior. The sex difference in the POA is achieved via estradiol's upregulation of the membrane-derived lipid signaling molecule prostaglandin E2 (PGE2); PGE2 is necessary and sufficient to masculinize both dendritic spine density and adult sexual behavior in rats. We have previously shown that PGE2 activates EP2 and EP4 receptors which increases protein kinase A (PKA) activity and that masculinized dendritic spine density and sex behavior are both dependent upon PKA as well as activation of AMPA type glutamate receptors. In the current experiments, we build upon this signaling cascade by determining that PGE2 induces phosphorylation of the AMPA receptor subunit, GluR1, which leads to increased AMPA receptor insertion at the membrane. Treating female pups on the day of birth with PGE2 induced the phosphorylation of GluR1 at the PKA-sensitive site within 2 hours of treatment, an effect that was blocked by co-administration of the PKA/AKAP inhibitor, HT31 with PGE2. Brief treatment of mixed neuronal/glial POA cultures with PGE2 or the cAMP/PKA stimulator, forskolin, increased membrane associated GluR1 in both neurons and glia. We speculate that PGE2 induced increases in AMPA receptor associated with the membrane underlies our previously observed increase in dendritic spine density and is a critical component in the masculinization of rodent sex behavior.
Collapse
Affiliation(s)
- Kathryn M Lenz
- Department of Physiology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, United States of America.
| | | | | | | |
Collapse
|
22
|
Ojeda SR, Lomniczi A, Sandau U. Contribution of glial-neuronal interactions to the neuroendocrine control of female puberty. Eur J Neurosci 2011; 32:2003-10. [PMID: 21143655 DOI: 10.1111/j.1460-9568.2010.07515.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mammalian puberty is initiated by an increased pulsatile release of the neuropeptide gonadotropin-releasing hormone (GnRH) from hypothalamic neuroendocrine neurons. Although this increase is primarily set in motion by neuronal networks synaptically connected to GnRH neurons, glial cells contribute to the process via at least two mechanisms. One involves production of growth factors acting via receptors endowed with either serine-threonine kinase or tyrosine kinase activity. The other involves plastic rearrangements of glia-GnRH neuron adhesiveness. Growth factors of the epidermal growth factor family acting via erbB receptors play a major role in glia-to-GnRH neuron communication. In turn, neurons facilitate astrocytic erbB signaling via glutamate-dependent cleavage of erbB ligand precursors. The genetic disruption of erbB receptors delays female sexual development due to impaired erbB ligand-induced glial prostaglandin E(2) release. The adhesiveness of glial cells to GnRH neurons involves at least two different cell-cell communication systems endowed with both adhesive and intracellular signaling capabilities. One is provided by synaptic cell adhesion molecule (SynCAM1), which establishes astrocyte-GnRH neuron adhesiveness via homophilic interactions and the other involves the heterophilic interaction of neuronal contactin with glial receptor-like protein tyrosine phosphatase-β. These findings indicate that the interaction of glial cells with GnRH neurons involves not only secreted bioactive molecules, but also cell-surface adhesive proteins able to set in motion intracellular signaling cascades.
Collapse
Affiliation(s)
- Sergio R Ojeda
- Division of Neuroscience, Oregon National Primate Research Center/Oregon Health Sciences University, 505 N.W., 185th Avenue, Beaverton, OR 97006, USA.
| | | | | |
Collapse
|
23
|
Ojeda SR, Lomniczi A, Loche A, Matagne V, Kaidar G, Sandau US, Dissen GA. The transcriptional control of female puberty. Brain Res 2010; 1364:164-74. [PMID: 20851111 PMCID: PMC2992593 DOI: 10.1016/j.brainres.2010.09.039] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 09/08/2010] [Accepted: 09/09/2010] [Indexed: 12/18/2022]
Abstract
The initiation of mammalian puberty requires a sustained increase in pulsatile release of gonadotrophin releasing hormone (GnRH) from the hypothalamus. This increase is brought about by coordinated changes in transsynaptic and glial-neuronal communication, consisting of an increase in neuronal and glial stimulatory inputs to the GnRH neuronal network and the loss of transsynaptic inhibitory influences. GnRH secretion is stimulated by transsynaptic inputs provided by excitatory amino acids (glutamate) and at least one peptide (kisspeptin), and by glial inputs provided by growth factors and small bioactive molecules. The inhibitory input to GnRH neurons is mostly transsynaptic and provided by GABAergic and opiatergic neurons; however, GABA has also been shown to directly excite GnRH neurons. There are many genes involved in the control of these cellular networks, and hence in the control of the pubertal process as a whole. Our laboratory has proposed the concept that these genes are arranged in overlapping networks internally organized in a hierarchical fashion. According to this concept, the highest level of intra-network control is provided by transcriptional regulators that, by directing expression of key subordinate genes, impose genetic coordination to the neuronal and glial subsets involved in initiating the pubertal process. More recently, we have begun to explore the concept that a more dynamic and encompassing level of integrative coordination is provided by epigenetic mechanisms.
Collapse
Affiliation(s)
- Sergio R Ojeda
- Division of Neuroscience, Oregon National Primate Research Center/Oregon Health and Science University, 505 N.W. 185th Avenue, Beaverton, OR 97006, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Prevot V, Hanchate NK, Bellefontaine N, Sharif A, Parkash J, Estrella C, Allet C, de Seranno S, Campagne C, de Tassigny XD, Baroncini M. Function-related structural plasticity of the GnRH system: a role for neuronal-glial-endothelial interactions. Front Neuroendocrinol 2010; 31:241-58. [PMID: 20546773 DOI: 10.1016/j.yfrne.2010.05.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 05/17/2010] [Accepted: 05/18/2010] [Indexed: 12/18/2022]
Abstract
As the final common pathway for the central control of gonadotropin secretion, GnRH neurons are subjected to numerous regulatory homeostatic and external factors to achieve levels of fertility appropriate to the organism. The GnRH system thus provides an excellent model in which to investigate the complex relationships between neurosecretion, morphological plasticity and the expression of a physiological function. Throughout the reproductive cycle beginning from postnatal sexual development and the onset of puberty to reproductive senescence, and even within the ovarian cycle itself, all levels of the GnRH system undergo morphological plasticity. This structural plasticity within the GnRH system appears crucial to the timely control of reproductive competence within the individual, and as such must have coordinated actions of multiple signals secreted from glial cells, endothelial cells, and GnRH neurons. Thus, the GnRH system must be viewed as a complete neuro-glial-vascular unit that works in concert to maintain the reproductive axis.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, F-59000 Lille, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sharif A, Prevot V. ErbB receptor signaling in astrocytes: a mediator of neuron-glia communication in the mature central nervous system. Neurochem Int 2010; 57:344-58. [PMID: 20685225 DOI: 10.1016/j.neuint.2010.05.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 03/29/2010] [Accepted: 05/18/2010] [Indexed: 10/19/2022]
Abstract
Astrocytes are now recognized as active players in the developing and mature central nervous system. Each astrocyte contacts vascular structures and thousands of synapses within discrete territories. These cells receive a myriad of inputs and generate appropriate responses to regulate the function of brain microdomains. Emerging evidence has implicated receptors of the ErbB tyrosine kinase family in the integration and processing of neuronal inputs by astrocytes: ErbB receptors can be activated by a wide range of neuronal stimuli; they control critical steps of glutamate-glutamine metabolism; and they regulate the biosynthesis and release of various glial-derived neurotrophic factors, gliomediators and gliotransmitters. These key properties of astrocytic ErbB signaling in neuron-glia interactions have significance for the physiology of the mature central nervous system, as exemplified by the central control of reproduction within the hypothalamus, and are also likely to contribute to pathological situations, since both dysregulation of ErbB signaling and glial dysfunction occur in many neurological disorders.
Collapse
Affiliation(s)
- Ariane Sharif
- Inserm, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the postnatal Brain, Lille, France.
| | | |
Collapse
|
26
|
Li B, Zhang S, Li M, Hertz L, Peng L. Serotonin increases ERK1/2 phosphorylation in astrocytes by stimulation of 5-HT2B and 5-HT2C receptors. Neurochem Int 2010; 57:432-9. [PMID: 20450948 DOI: 10.1016/j.neuint.2010.04.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Revised: 03/24/2010] [Accepted: 04/26/2010] [Indexed: 11/29/2022]
Abstract
We have previously shown that fluoxetine causes ERK(1/2) phosphorylation in cultured mouse astrocytes mediated exclusively by stimulation of 5-HT(2B) receptors (Li et al., 2008b). This raises the question whether this is also the case for serotonin (5-HT) itself. In the present study serotonin was found to induce ERK(1/2) phosphorylation by stimulation of 5-HT(2B) receptors with high affinity (EC(50): 20-30 pM), and by stimulation of 5-HT(2C) receptor with low affinity (EC(50): 1 microM or higher). ERK(1/2) phosphorylation induced by stimulation of either 5-HT(2B) or 5-HT(2C) receptors was mediated by epidermal growth factor (EGF) receptor transactivation (Peng et al., this issue), shown by the inhibitory effect of AG1478, an inhibitor of the EGF receptor tyrosine kinase, and GM6001, an inhibitor of Zn-dependent metalloproteinases, and thus of 5-HT(2B) receptor-mediated EGF receptor agonist release. It is discussed that the high potency of the 5-HT(2B)-mediated effect is consistent with literature data for binding affinity of serotonin to cloned human 5-HT(2B) receptors and with observations of low extracellular concentrations of serotonin in brain, which would allow a demonstrated moderate and modality-dependent increase in specific brain areas to activate 5-HT(2B) receptors. In contrast the relevance of the observed 5-HT(2C) receptors on astrocytes is questioned.
Collapse
Affiliation(s)
- Baoman Li
- Department of Clinical Pharmacology, China Medical University, Heping District, Shenyang, PR China
| | | | | | | | | |
Collapse
|
27
|
Glutamate Promotes Cell Growth by EGFR Signaling on U-87MG Human Glioblastoma Cell Line. Pathol Oncol Res 2009; 16:285-93. [DOI: 10.1007/s12253-009-9223-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Accepted: 10/20/2009] [Indexed: 10/20/2022]
|
28
|
Yin W, Wu D, Noel ML, Gore AC. Gonadotropin-releasing hormone neuroterminals and their microenvironment in the median eminence: effects of aging and estradiol treatment. Endocrinology 2009; 150:5498-508. [PMID: 19819960 PMCID: PMC2795719 DOI: 10.1210/en.2009-0679] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The GnRH decapeptide controls reproductive function through its release from neuroendocrine terminals in the median eminence, a site where there is a convergence of numerous nerve terminals and glial cells. Previous work showed dynamic changes in the GnRH-glial-capillary network in the median eminence under different physiological conditions. Because aging in rats is associated with a diminution of GnRH release and responsiveness to estradiol feedback, we examined effects of age and estradiol treatment on these anatomical interactions. Rats were ovariectomized at young (4 months), middle-aged (11 months), or old (22-23 months) ages, allowed 4 wk to recover, and then treated with vehicle or estradiol for 72 h followed by perfusion. Immunofluorescence of GnRH was measured, and immunogold electron microscopic analyses were performed to study the ultrastructural properties of GnRH neuroterminals and their microenvironment. Although the GnRH immunofluorescent signal showed no significant changes with age and estradiol treatment, we found that the median eminence underwent both qualitative and quantitative structural changes with age, including a disorganization of cytoarchitecture with aging and a decrease in the apposition of GnRH neuroterminals to glia with age and estradiol treatment. Thus, although GnRH neurons can continue to synthesize and transport peptide, changes in the GnRH neuroterminal-glial-capillary machinery occur during reproductive senescence in a manner consistent with a disconnection of these elements and a potential dysregulation of GnRH neurosecretion.
Collapse
Affiliation(s)
- Weiling Yin
- Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | | |
Collapse
|
29
|
ERK phosphorylation in intact, adult brain by α2-adrenergic transactivation of EGF receptors. Neurochem Int 2009; 55:593-600. [DOI: 10.1016/j.neuint.2009.05.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2008] [Revised: 05/27/2009] [Accepted: 05/28/2009] [Indexed: 11/23/2022]
|
30
|
Sharif A, Duhem-Tonnelle V, Allet C, Baroncini M, Loyens A, Kerr-Conte J, Collier F, Blond S, Ojeda SR, Junier MP, Prevot V. Differential erbB signaling in astrocytes from the cerebral cortex and the hypothalamus of the human brain. Glia 2009; 57:362-79. [DOI: 10.1002/glia.20762] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
31
|
Johnson RT, Breedlove SM, Jordan CL. Sex differences and laterality in astrocyte number and complexity in the adult rat medial amygdala. J Comp Neurol 2008; 511:599-609. [PMID: 18853427 PMCID: PMC2592304 DOI: 10.1002/cne.21859] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The posterodorsal portion of the medial amygdala (MePD) is sexually dimorphic in several rodent species. In several other brain nuclei, astrocytes change morphology in response to steroid hormones. We visualized MePD astrocytes using glial-fibrillary acidic protein (GFAP) immunocytochemistry. We compared the number and process complexity of MePD astrocytes in adult wildtype male and female rats and testicular feminized mutant (TFM) male rats that lack functional androgen receptors (ARs) to determine whether MePD astrocytes are sexually differentiated and whether ARs have a role. Unbiased stereological methods revealed laterality and sex differences in MePD astrocyte number and complexity. The right MePD contained more astrocytes than the left in all three genotypes, and the number of astrocytes was also sexually differentiated in the right MePD, with males having more astrocytes than females. In contrast, the left MePD contained more complex astrocytes than did the right MePD in all three genotypes, and males had more complex astrocytes than females in this hemisphere. TFM males were comparable to wildtype females, having fewer astrocytes on the right and simpler astrocytes on the left than do wildtype males. Taken together, these results demonstrate that astrocytes are sexually dimorphic in the adult MePD and that the nature of the sex difference is hemisphere-dependent: a sex difference in astrocyte number in the right MePD and a sex difference in astrocyte complexity in the left MePD. Moreover, functional ARs appear to be critical in establishing these sex differences in MePD astrocyte morphology.
Collapse
Affiliation(s)
- Ryan T Johnson
- Neuroscience Program, Michigan State University, East Lansing, Michigan 48824-1101, USA.
| | | | | |
Collapse
|
32
|
Surena AL, de Faria GP, Studler JM, Peiretti F, Pidoux M, Camonis J, Chneiweiss H, Formstecher E, Junier MP. DLG1/SAP97 modulates transforming growth factor alpha bioavailability. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:264-72. [PMID: 18930083 DOI: 10.1016/j.bbamcr.2008.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Revised: 08/06/2008] [Accepted: 09/11/2008] [Indexed: 11/27/2022]
Abstract
TGFalpha and its receptor EGFR participate in the development of a wide range of tumors including gliomas, the main adult primary brain tumors. TGFalpha soluble form results from the cleavage by the metalloprotease TACE/ADAM17 of the extracellular part of its transmembrane precursor, pro-TGFalpha. To gain insights into the mechanisms underlying TGFalpha bioavailability, a yeast two-hybrid screen was performed to identify proteins interacting with pro-TGFalpha intracellular domain (ICD). DLG1/SAP97 (Discs Large Gene 1 or Synapse Associated Protein 97) was found to interact with both pro-TGFalpha and TACE ICDs through distinct PDZ domains. An in vivo pro-TGFalpha-DLG1-TACE complex was detected in U251 glioma cells and in gliomas-derived tumor initiating cells. Interaction between DLG1 and TACE diminished in response to stimulations promoting pro-TGFalpha shedding. Manipulation of DLG1 levels revealed dual actions of DLG1 on pro-TGFalpha shedding, favoring approximation of pro-TGFalpha and TACE, while limiting TACE full shedding activity. These results show that DLG1 participates in the control of TGFalpha bioavailability through its dynamic interaction with the growth factor precursor and TACE.
Collapse
|
33
|
Durand D, Pampillo M, Caruso C, Lasaga M. Role of metabotropic glutamate receptors in the control of neuroendocrine function. Neuropharmacology 2008; 55:577-83. [PMID: 18616955 DOI: 10.1016/j.neuropharm.2008.06.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 05/23/2008] [Accepted: 06/12/2008] [Indexed: 11/25/2022]
Abstract
Glutamate exerts its effects through binding and activation of two classes of specific receptors: ionotropic (iGluRs) and metabotropic (mGluRs). Group I mGluR includes mGluR1 and mGluR5 subtypes, group II includes mGluR2 and mGluR3 subtypes and group III includes the subtypes mGluR 4, 6, 7 and 8. Glutamate and its receptors are found in all key hypothalamic areas critically involved in reproduction and neuroendocrine function. To date, considerable data support an important role for iGluRs in the control of neuroendocrine function; however, the role of mGluRs as regulators of hypothalamic-pituitary function has not been clearly elucidated. mGluRs could be exerting a fine tune on the release of hypothalamic factors that regulate hormone release such as Substance P, GABA, alpha-MSH and CRH. Group II mGluR exert a direct inhibitory effect on anterior pituitary prolactin and GH secretion. Moreover, some group II mGluR agonists, like LY 354,740 and LY 379,268, can modulate PRL secretion from the anterior pituitary through their actions as dopamine receptor agonists. Evidence suggests a role for group III mGluR subtypes in stress-related behavioral disorders. Several reports indicate that selective ligands for mGluR subtypes have potential for the treatment of a wide variety of neurological and psychiatric disorders, including depression, anxiety disorders, schizophrenia, epilepsy and Alzheimer's disease among others. Since converging lines of evidence suggest a role for mGluRs subtypes in neuroendocrine regulation of hormone secretion, mGluRs neuroendocrine actions must be taken in consideration to insure proper treatment of these diseases. Moreover, discovery of selective agonists provides an opportunity to investigate the physiological role of mGluR subtypes and to directly test the neuroendocrine actions of mGluRs. Finally, mGluRs selective agonists may have an impact in the treatment of conditions involving chronic stress, such as depression and anxiety disorders, since they regulate neuroendocrine stress circuits involving the HPA axis and stress-sensitive hormones such as oxytocin and prolactin. This review aims to provide a survey of our current understanding of the effects of mGluR activation on neuroendocrine function.
Collapse
Affiliation(s)
- Daniela Durand
- Research Center in Reproduction, School of Medicine, University of Buenos Aires, Paraguay 2155, Piso 10, 1121 Buenos Aires, Argentina
| | | | | | | |
Collapse
|
34
|
Ojeda SR, Lomniczi A, Sandau US. Glial-gonadotrophin hormone (GnRH) neurone interactions in the median eminence and the control of GnRH secretion. J Neuroendocrinol 2008; 20:732-42. [PMID: 18601696 DOI: 10.1111/j.1365-2826.2008.01712.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A wealth of information now exists showing that glial cells are actively involved in the cell-cell communication process generating and disseminating information within the central nervous system. In the hypothalamus, two types of glial cells, astrocytes and ependymal cells lining the latero-ventral portion of the third ventricle (known as tanycytes), regulate the secretory activity of neuroendocrine neurones. This function, initially described for astrocytes apposing magnocellular neurones, has been more recently characterised for neurones secreting gonadotrophin hormone-releasing hormone (GnRH). The available evidence suggests that glial cells of the median eminence regulate GnRH secretion via two related mechanisms. One involves the production of growth factors acting via receptors with tyrosine kinase activity. The other involves plastic rearrangements of glia-GnRH neurone adhesiveness. GnRH axons reach the median eminence, at least in part, directed by basic fibroblast growth factor. Their secretory activity is facilitated by insulin-like growth factor 1 and members of the epidermal growth factor family. A structural complement to these soluble molecules is provided by at least three cell-cell adhesion systems endowed with signalling capabilities. One of them uses the neuronal cell adhesion molecule (NCAM), another employs the synaptic cell adhesion molecule (SynCAM), and the third one consists of neuronal contactin interacting with glial receptor-like protein tyrosine phosphatase-beta. It is envisioned that, within the median eminence, soluble factors and adhesion molecules work coordinately to control delivery of GnRH to the portal vasculature.
Collapse
Affiliation(s)
- S R Ojeda
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | | | | |
Collapse
|
35
|
Schneider MR, Wolf E. The epidermal growth factor receptor and its ligands in female reproduction: Insights from rodent models. Cytokine Growth Factor Rev 2008; 19:173-81. [DOI: 10.1016/j.cytogfr.2008.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Prevot V, Dehouck B, Poulain P, Beauvillain JC, Buée-Scherrer V, Bouret S. Neuronal-glial-endothelial interactions and cell plasticity in the postnatal hypothalamus: implications for the neuroendocrine control of reproduction. Psychoneuroendocrinology 2007; 32 Suppl 1:S46-51. [PMID: 17629628 DOI: 10.1016/j.psyneuen.2007.03.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Revised: 03/06/2007] [Accepted: 03/13/2007] [Indexed: 10/23/2022]
Abstract
It is becoming increasingly apparent that non-neuronal cells play a critical role in generating and regulating the flow of information within the brain. Among these non-neuronal cells, astroglial cells have been shown to play important roles in the control of both synaptic transmission and neurosecretion. In addition to modulating neuronal activity, astroglial cells interact with endothelial cells throughout the central nervous system to define specific functional domains. In the hypothalamus, neurons that release gonadotropin-releasing hormone (GnRH), the neurohormone that controls both sexual development and adult reproductive function, offer an attractive model system in which to study glial-neuronal-endothelial interactions. Within the median eminence of the hypothalamus, alterations of the anatomical relationship that exists between GnRH axon terminals and ependymoglial cell processes belonging to tanycytes regulate the direct access of GnRH neurosecretory axons to the vascular wall. This cell plasticity presumably modulates the release of GnRH into the portal vasculature during the reproductive cycle. Both structural changes and GnRH secretory activity appear to be modulated, at least in part, by specific cell-cell signalling molecules secreted by astrocytes, tanycytes and endothelial cells. It is becoming increasingly clear that among the different factors that may be involved, glial cells use growth factor members of the epidermal growth factor (EGF) family, acting via receptors endowed with tyrosine kinase activity, to produce morphological changes and release neuroactive substances that directly excite nearby neurons, whereas endothelial cells of the median eminence employ nitric oxide to induce neuroglial plasticity and facilitate GnRH release.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Jean-Pierre Aubert Research Centre, U837, Development and Plasticity of the Postnatal Brain, Place de Verdun, 59045 Lille Cedex, France.
| | | | | | | | | | | |
Collapse
|
37
|
Roth CL, McCormack AL, Lomniczi A, Mungenast AE, Ojeda SR. Quantitative proteomics identifies a change in glial glutamate metabolism at the time of female puberty. Mol Cell Endocrinol 2006; 254-255:51-9. [PMID: 16753258 DOI: 10.1016/j.mce.2006.04.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mammalian puberty requires activation of luteinizing hormone-releasing hormone (LHRH) neurons. In turn, these neurons are controlled by transsynaptic and glia-to-neuron communication pathways, which employ diverse cellular proteins for proper function. We have now used a high throughput relative quantitative proteomics technique to identify such proteins. We selected the method of two-dimensional liquid chromatography tandem mass spectrometry (2DLC-MS/MS) and cleavable isotope-coded affinity tags (cICAT), to both identify and quantify individual proteins within a complex protein mixture. The proteins used derived from the hypothalamus of juvenile (25-day-old) and peripubertal (first proestrus, LP) female rats, and their identity was established by analyzing their mass spectra via database searching. Five proteins involved in glutamate metabolism were detected and two of them appeared to be differentially expressed. They were selected for further analysis, because of their importance in controlling glutamate synthesis and degradation, and their preferential expression in astroglial cells. One, glutamate dehydrogenase (GDH) catalyzes glutamate synthesis; its hypothalamic content detected by 2DLC-MS/MS increases at first proestrus. The other, glutamine synthetase (GS), catalyzes the metabolism of glutamate to glutamine; its content decreases in proestrus. Western blot analysis verified these results. Because these changes suggested an increased glutamate production at puberty, we measured glutamate release from hypothalamic fragments from juvenile 29-day old rats, and from rats treated with PMSG to induce a premature proestrus surge of luteinizing hormone (LH). To determine the net output of glutamate in the absence of re-uptake we used the excitatory amino acid transporter (EAAT) inhibitor l-trans-pyrrolidine-2,4-dicarboxylic acid (PDC). PDC elicited significantly more glutamate- and LHRH-release from the proestrus hypothalamus. Thus, an increase excitatory drive to the LHRH neuronal network provided by glutamatergic inputs of glial origin, is an event contributing to the pubertal activation of LHRH secretion.
Collapse
Affiliation(s)
- Christian L Roth
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | | | | | | | | |
Collapse
|
38
|
Ojeda SR, Roth C, Mungenast A, Heger S, Mastronardi C, Parent AS, Lomniczi A, Jung H. Neuroendocrine mechanisms controlling female puberty: new approaches, new concepts. ACTA ACUST UNITED AC 2006; 29:256-63; discussion 286-90. [PMID: 16466547 DOI: 10.1111/j.1365-2605.2005.00619.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sexual development and mature reproductive function are controlled by a handful of neurones that, located in the basal forebrain, produce the decapeptide luteinizing hormone releasing hormone (LHRH). LHRH is released into the portal system that connects the hypothalamus to the pituitary gland and act on the latter to stimulate the synthesis and release of gonadotrophin hormones. The pubertal activation of LHRH release requires coordinated changes in excitatory and inhibitory inputs to LHRH-secreting neurones. These inputs are provided by both transsynaptic and glia-to-neurone communication pathways. Using cellular and molecular approaches, in combination with transgenic animal models and high-throughput procedures for gene discovery, we are gaining new insight into the basic mechanisms underlying this dual control of LHRH secretion and, hence, the initiation of mammalian puberty. Our results suggest that the initiation of puberty requires reciprocal neurone-glia communication involving excitatory amino acids and growth factors, and the coordinated actions of a group of transcriptional regulators that appear to represent a higher level of control governing the pubertal process.
Collapse
Affiliation(s)
- Sergio R Ojeda
- Division of Neuroscience, Oregon National Primate Research Center/Oregon, Health & Science University, Beaverton, 97006, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Ojeda SR, Lomniczi A, Mastronardi C, Heger S, Roth C, Parent AS, Matagne V, Mungenast AE. Minireview: the neuroendocrine regulation of puberty: is the time ripe for a systems biology approach? Endocrinology 2006; 147:1166-74. [PMID: 16373420 DOI: 10.1210/en.2005-1136] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The initiation of mammalian puberty requires an increase in pulsatile release of GnRH from the hypothalamus. This increase is brought about by coordinated changes in transsynaptic and glial-neuronal communication. As the neuronal and glial excitatory inputs to the GnRH neuronal network increase, the transsynaptic inhibitory tone decreases, leading to the pubertal activation of GnRH secretion. The excitatory neuronal systems most prevalently involved in this process use glutamate and the peptide kisspeptin for neurotransmission/neuromodulation, whereas the most important inhibitory inputs are provided by gamma-aminobutyric acid (GABA)ergic and opiatergic neurons. Glial cells, on the other hand, facilitate GnRH secretion via growth factor-dependent cell-cell signaling. Coordination of this regulatory neuronal-glial network may require a hierarchical arrangement. One level of coordination appears to be provided by a host of unrelated genes encoding proteins required for cell-cell communication. A second, but overlapping, level might be provided by a second tier of genes engaged in specific cell functions required for productive cell-cell interaction. A third and higher level of control involves the transcriptional regulation of these subordinate genes by a handful of upper echelon genes that, operating within the different neuronal and glial subsets required for the initiation of the pubertal process, sustain the functional integration of the network. The existence of functionally connected genes controlling the pubertal process is consistent with the concept that puberty is under genetic control and that the genetic underpinnings of both normal and deranged puberty are polygenic rather than specified by a single gene. The availability of improved high-throughput techniques and computational methods for global analysis of mRNAs and proteins will allow us to not only initiate the systematic identification of the different components of this neuroendocrine network but also to define their functional interactions.
Collapse
Affiliation(s)
- Sergio R Ojeda
- Division of Neuroscience, Oregon National Primate Research Center/Oregon Health and Science University, 505 Northwest 185th Avenue, Beaverton, Oregon 97006, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Parent AS, Matagne V, Bourguignon JP. Control of puberty by excitatory amino acid neurotransmitters and its clinical implications. Endocrine 2005; 28:281-6. [PMID: 16388117 DOI: 10.1385/endo:28:3:281] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2005] [Accepted: 07/13/2005] [Indexed: 11/11/2022]
Abstract
Excitatory amino acids, glutamate in particular, have a marked stimulatory effect on the reproductive axis, particularly at puberty. Glutamate, N-methyl-D-aspartate (NMDA), and kainate stimulate gonadotropin-releasing hormone (GnRH) secretion in immature mammals and NMDA receptor stimulation results in precocious puberty in rats and monkeys. Puberty is characterized by an increased sensitivity of GnRH to glutamate as well as an increase in glutaminase activity in the hypothalamus. Glutamatergic and GABAergic regulation of GnRH secretion seem strongly interdependent around puberty. In addition to the transsynaptic glutamatergic regulation of GnRH secretion, a coordinated activity of glutamatergic neurons and astroglial cells has been shown to play an active role in puberty. The participation of kainate receptors in the estradiol-induced advancement of puberty suggest that these receptors may be involved in the estradiol-mediated activation of GnRH secretion at puberty. A case of precocious puberty associated with hyperglycinemia illustrates the NMDA involvement in puberty in humans. In this patient, the occurrence of precocious puberty was thought to result from excessive stimulation by glycine of the NMDA receptors linked to the GnRH neurons. Glutamate plays several roles in the hypothalamic mechanism of puberty as it has been shown in animal models, but there are still few clinical data supporting the role of glutamate in human puberty.
Collapse
Affiliation(s)
- Anne-Simone Parent
- Developmental Neuroendocrinology, Center for Molecular and Cellular Neurobiology, University of Liège, Belgium
| | | | | |
Collapse
|
41
|
Volterra A, Meldolesi J. Astrocytes, from brain glue to communication elements: the revolution continues. Nat Rev Neurosci 2005; 6:626-40. [PMID: 16025096 DOI: 10.1038/nrn1722] [Citation(s) in RCA: 1259] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
For decades, astrocytes have been considered to be non-excitable support cells of the brain. However, this view has changed radically during the past twenty years. The recent recognition that they are organized in separate territories and possess active properties--notably a competence for the regulated release of 'gliotransmitters', including glutamate--has enabled us to develop an understanding of previously unknown functions for astrocytes. Today, astrocytes are seen as local communication elements of the brain that can generate various regulatory signals and bridge structures (from neuronal to vascular) and networks that are otherwise disconnected from each other. Examples of their specific and essential roles in normal physiological processes have begun to accumulate, and the number of diseases known to involve defective astrocytes is increasing.
Collapse
Affiliation(s)
- Andrea Volterra
- Department of Cell Biology and Morphology, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland.
| | | |
Collapse
|
42
|
Klugmann M, Symes CW, Leichtlein CB, Klaussner BK, Dunning J, Fong D, Young D, During MJ. AAV-mediated hippocampal expression of short and long Homer 1 proteins differentially affect cognition and seizure activity in adult rats. Mol Cell Neurosci 2005; 28:347-60. [PMID: 15691715 DOI: 10.1016/j.mcn.2004.10.002] [Citation(s) in RCA: 164] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2004] [Revised: 10/01/2004] [Accepted: 10/01/2004] [Indexed: 01/17/2023] Open
Abstract
Homer proteins mediate molecular rearrangements leading to changes in spine morphology. This points to a role of Homer in learning and memory. Homer 1c features both the ligand binding domain and a coiled-coiled domain for self-multimerization. Homer 1a lacks the coiled-coiled domain. Here, we report a new isoform which we termed 1g, lacking the Homer ligand binding domain. We dissected the functional roles of the individual Homer 1 domains, encoded by Homer 1a, 1c, and 1g, in vivo. Recombinant adeno-associated virus (AAV)-mediated overexpression of these forms in the hippocampus of adult rats has opposing effects on learning behavior. Increased levels of Homer 1a impaired hippocampal-dependent memory, while Homer 1g and 1c slightly enhanced memory performance. Homer 1g induced anxiety. Moreover, AAV-Homer 1a animals showed attenuation of electrographic seizures in a model of status epilepticus. These results suggest that Homer 1 proteins play an active role in behavioral plasticity.
Collapse
Affiliation(s)
- Matthias Klugmann
- Department of Molecular Medicine and Pathology, Functional Genomics and Translational Neuroscience Laboratory, University of Auckland, Auckland, New Zealand.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Palmer CL, Cotton L, Henley JM. The molecular pharmacology and cell biology of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. Pharmacol Rev 2005; 57:253-77. [PMID: 15914469 PMCID: PMC3314513 DOI: 10.1124/pr.57.2.7] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptors (AMPARs) are of fundamental importance in the brain. They are responsible for the majority of fast excitatory synaptic transmission, and their overactivation is potently excitotoxic. Recent findings have implicated AMPARs in synapse formation and stabilization, and regulation of functional AMPARs is the principal mechanism underlying synaptic plasticity. Changes in AMPAR activity have been described in the pathology of numerous diseases, such as Alzheimer's disease, stroke, and epilepsy. Unsurprisingly, the developmental and activity-dependent changes in the functional synaptic expression of these receptors are under tight cellular regulation. The molecular and cellular mechanisms that control the postsynaptic insertion, arrangement, and lifetime of surface-expressed AMPARs are the subject of intense and widespread investigation. For example, there has been an explosion of information about proteins that interact with AMPAR subunits, and these interactors are beginning to provide real insight into the molecular and cellular mechanisms underlying the cell biology of AMPARs. As a result, there has been considerable progress in this field, and the aim of this review is to provide an account of the current state of knowledge.
Collapse
Affiliation(s)
- Claire L Palmer
- Medical Research Council Centre for Synaptic Plasticity, Department of Anatomy, School of Medical Sciences, Bristol University, Bristol, UK
| | | | | |
Collapse
|
44
|
Mungenast AE, Ojeda SR. Expression of three gene families encoding cell-cell communication molecules in the prepubertal nonhuman primate hypothalamus. J Neuroendocrinol 2005; 17:208-19. [PMID: 15842232 DOI: 10.1111/j.1365-2826.2005.01293.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Transsynaptic and glial-neuronal communication are important components of the mechanism underlying the pubertal activation of luteinizing hormone-releasing hormone (LHRH) secretion. The molecules required for the architectural organization of these cell-cell interactions have not been identified. We now show that the hypothalamus of the prepubertal female rhesus monkey expresses a multiplicity of genes encoding three families of adhesion/signalling proteins involved in the structural definition of both neurone-to-neurone and bi-directional neurone-glia communication. These include the neurexin/neuroligin (NRX/NRL) and protocadherin-alpha (PCDHalpha) families of synaptic specifiers/adhesion molecules, and key components of the contactin-dependent neuronal-glial adhesiveness complex, including contactin/F3 itself, the contactin-associated protein-1 (CASPR1), and the glial receptor protein tyrosine phosphatase beta. Prominently expressed among members of the NRX family is the neurexin isoform involved in the specification of glutamatergic synapses. Although NRXs, PCDHalphas and CASPR1 transcripts are mostly detected in neurones, the topography of expression appears different. NRX1 mRNA-containing neurones are scattered throughout the hypothalamus, PCDHalpha mRNA transcripts appear more abundant in neurones of the arcuate nucleus and periventricular region, and neurones positive for CASPR1 mRNA exhibit a particularly striking distribution pattern that delineates the hypothalamus. Examination of LHRH neurones, using the LHRH-secreting cell line GT1-7, showed that these cells contain transcripts encoding NRXs and one of their ligands (NRL1), at least one PCDHalpha (CNR-8/PCDHalpha10), and the CASPR1/contactin complex. The results indicate that the prepubertal female monkey hypothalamus contains a plethora of adhesion/signalling molecules with different but complementary functions, and that an LHRH neuronal cell line expresses key components of this structural complex. The presence of such cell-cell communication machinery in the neuroendocrine brain suggests an integrated participation of their individual components in the central control of female sexual development.
Collapse
Affiliation(s)
- A E Mungenast
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR, USA
| | | |
Collapse
|
45
|
Prevot V, Lomniczi A, Corfas G, Ojeda SR. erbB-1 and erbB-4 receptors act in concert to facilitate female sexual development and mature reproductive function. Endocrinology 2005; 146:1465-72. [PMID: 15591145 DOI: 10.1210/en.2004-1146] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Glial erbB-1 and erbB-4 receptors are key components of the process by which neuroendocrine glial cells control LHRH secretion and the onset of female puberty. We now provide evidence that these two signaling systems work in a coordinated fashion to control reproductive function. To generate animals carrying functionally impaired erbB-1 and erbB-4 receptors, we crossed Waved 2 (Wa-2+/+) mice harboring a point mutation of the erbB-1 receptor with mice expressing a dominant-negative erbB-4 receptor in astrocytes. In comparison to single-deficient mice, double-mutant animals exhibited a further delay in the onset of puberty and a strikingly diminished adult reproductive capacity. Ligand-dependent erbB receptor phosphorylation and erbB-mediated MAPK (ERK 1/2) phosphorylation were impaired in mutant astrocytes. Wa-2+/+ or double-mutant astrocytes failed to respond to TGF alpha with production of prostaglandin E2, one of the factors mediating the stimulatory effect of astroglial erbB receptor activation on LHRH release. Medium conditioned by Wa-2+/+ or double-mutant astrocytes treated with TGF alpha failed to stimulate LHRH release from GT1-7 cells. The LH response to ovariectomy was significantly attenuated in mutant mice in comparison with wild-type controls. Although the Wa-2 mutation affects all cells bearing erbB-1 receptors, these results suggest that a major defect underlying the reproductive defects of animals with impaired erbB signaling is a decreased ability of glial cells to stimulate LHRH release. Thus, a coordinated involvement of erbB-1 and erbB-4 signaling systems is required for the normalcy of sexual development and the maintenance of mature female reproductive function.
Collapse
Affiliation(s)
- Vincent Prevot
- Division of Neuroscience, Oregon National Primate Research Center/Oregon Health & Sciences Unversity, 505 Northwest 185th Avenue, Beaverton, Oregon 97006, USA
| | | | | | | |
Collapse
|
46
|
Eyigor O, Minbay Z, Cavusoglu I, Jennes L. Localization of kainate receptor subunit GluR5-immunoreactive cells in the rat hypothalamus. ACTA ACUST UNITED AC 2005; 136:38-44. [PMID: 15893585 DOI: 10.1016/j.molbrainres.2005.01.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2004] [Revised: 12/22/2004] [Accepted: 01/01/2005] [Indexed: 11/18/2022]
Abstract
Glutamate is the major excitatory neurotransmitter in the hypothalamus, which exerts its effects by activating ion channel-forming (ionotropic) or G-protein-coupled (metabotropic) receptors. Kainate-preferring glutamate receptor subunits (GluR5, GluR6, GluR7, KA1, and KA2) form one of the three ionotropic receptor families. In the present study, we analyzed the distribution of GluR5 subunit protein in the rat hypothalamus with immunohistochemistry. GluR5 immunoreactivity was observed in perikarya and processes of many hypothalamic cells some of which, based upon their morphological differentiation by size and structure, appeared to be neurons and others glial cells. Analyses revealed that higher number of glial cells were GluR5 positive when compared to the moderate number of GluR5-labeled neurons in the anteroventral periventricular nucleus. Numerous GluR5-expressing neurons and similar number of glia were detected in the suprachiasmatic nucleus. In the arcuate nucleus more glial cells were identified with GluR5 immunoreactivity than the number of labeled neurons. Scattered GluR5-positive cells were present in the periventricular nucleus. Specific immunostaining was not seen in the ventromedial nucleus or dorsomedial nucleus. In conclusion, it is suggested that the GluR5 subunits participate in the glutamatergic regulation of several neuroendocrine systems, such as the tubero-infundibular systems as well as in the control of circadian output through neuron-to-neuron and/or neuron-to-glia interactions.
Collapse
Affiliation(s)
- Ozhan Eyigor
- Department of Histology and Embryology, Uludag University, Faculty of Medicine, Gorukle Kampusu, 16059, Bursa, Turkey.
| | | | | | | |
Collapse
|
47
|
Abstract
Modulating protein-protein interactions involved in disease pathways is an attractive strategy for developing drugs, but remains a challenge to achieve. One approach is to target certain domains within proteins that mediate these interactions. One example of such a domain is the PDZ domain, which is involved in interactions between many different proteins in a variety of cellular contexts. Because PDZ domains have well-defined binding sites, they are promising targets for drug discovery. However, there is still much to learn about the function of these domains before drugs targeting PDZ interactions can become a reality.
Collapse
Affiliation(s)
- Kumlesh K Dev
- Neuroscience Research, Novartis Institutes for BioMedical Research, Novartis Pharma AG, WSJ-386.7.43, CH-4002 Basel, Switzerland.
| |
Collapse
|
48
|
Bruzzone S, Verderio C, Schenk U, Fedele E, Zocchi E, Matteoli M, De Flora A. Glutamate-mediated overexpression of CD38 in astrocytes cultured with neurones. J Neurochem 2004; 89:264-72. [PMID: 15030411 DOI: 10.1111/j.1471-4159.2003.02326.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Recently, a new system of astrocyte-neurone glutamatergic signalling has been identified. It is started in astrocytes by ectocellular, CD38-catalysed conversion of NAD(+) to the calcium mobilizer cyclic ADP-ribose (cADPR). This is then pumped by CD38 itself into the cytosol where the resulting free intracellular Ca(2+) concentration [Ca(2+)](i) transients elicit an increased release of glutamate, which can induce an enhanced Ca(2+) response in neighbouring neurones. Here, we demonstrate that co-culture of either cortical or hippocampal astrocytes with neurones results in a significant overexpression of astrocyte CD38 both on the plasma membrane and intracellularly. The causal role of neurone-released glutamate in inducing overexpression of astrocyte CD38 is demonstrated by two observations: first, in the absence of neurones, induction of CD38 in pure astrocyte cultures can be obtained with glutamate and second, it can be prevented in co-cultures by glutamate receptor antagonists. The neuronal glutamate-mediated effect of neurones on astrocyte CD38 expression is paralleled by increased intracellular cADPR and [Ca(2+)](i) levels, both findings indicating functionality of overexpressed CD38. These results reveal a new neurone-to-astrocyte glutamatergic signalling based on the CD38/cADPR system, which affects the [Ca(2+)](i) in both cell types, adding further complexity to the bi-directional patterns of communication between astrocytes and neurones.
Collapse
Affiliation(s)
- Santina Bruzzone
- Department of Experimental Medicine, Sections of Biochemistry and Pharmacology, University of Genova, Italy
| | | | | | | | | | | | | |
Collapse
|
49
|
Shah BH, Catt KJ. GPCR-mediated transactivation of RTKs in the CNS: mechanisms and consequences. Trends Neurosci 2004; 27:48-53. [PMID: 14698610 DOI: 10.1016/j.tins.2003.11.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Bukhtiar H Shah
- Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-4510, USA
| | | |
Collapse
|
50
|
Prevot V, Cornea A, Mungenast A, Smiley G, Ojeda SR. Activation of erbB-1 signaling in tanycytes of the median eminence stimulates transforming growth factor beta1 release via prostaglandin E2 production and induces cell plasticity. J Neurosci 2003; 23:10622-32. [PMID: 14627647 PMCID: PMC6740908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023] Open
Abstract
The activation of transforming growth factor alpha (TGFalpha)-erbB-1 and neuregulin-erbB-4 signaling pathways in hypothalamic astrocytes has been shown to play a key role in the process by which the neuroendocrine brain controls luteinizing hormone-releasing hormone (LHRH) secretion. Earlier studies suggested that tanycytes, an ependymoglial cell type of the median eminence, regulate LHRH release during the estrous cycle by undergoing plastic changes that alternatively allow or prevent direct access of the LHRH nerve terminals to the portal vasculature. Neither the molecules responsible for these plastic changes nor the underlying controlling mechanisms have been identified. Here we show that cultured tanycytes express erbB-1 and erbB-2, two of the four members of the erbB receptor family, and respond to TGFalpha with receptor phosphorylation, release of prostaglandin E2 (PGE2), and a PGE2-dependent increase in the release of TGFbeta1, a growth factor previously implicated in the glial control of LHRH secretion. Blockade of either erbB-1 receptor signal transduction or prostaglandin synthesis prevented the stimulatory effect of TGFalpha on both PGE2 and TGFbeta1 release. Time-lapse studies revealed that TGFalpha and TGFbeta1 have dramatically opposite effects on tanycyte plasticity. Whereas TGFalpha promotes tanycytic outgrowth, TGFbeta1 elicits retraction of tanycytic processes. Blockade of metalloproteinase activity abolished the effect of TGFbeta1, suggesting that TGFbeta1 induces tanycytic retraction by facilitating dissolution of the extracellular matrix. Prolonged (>12 hr) exposure of tanycytes to TGFalpha resulted in focal tanycytic retraction, an effect that was abolished by immunoneutralization of TGFbeta1 action, indicating that the retraction was attributable to TGFalpha-induced TGFbeta1 formation. These in vitro results identify tanycytes as targets of TGFalpha action and demonstrate that activation of erbB-1-mediated signaling in these cells results in plastic changes that, involving PGE2 and TGFbeta1 as downstream effectors, mimic the morphological plasticity displayed by tanycytes during the hours encompassing the preovulatory surge of LHRH.
Collapse
Affiliation(s)
- Vincent Prevot
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon 97006, USA
| | | | | | | | | |
Collapse
|