1
|
Buckley M, Jacob WP, Bortey L, McClain ME, Ritter AL, Godfrey A, Munneke AS, Ramachandran S, Kenis S, Kolnik JC, Olofsson S, Nenadovich M, Kutoloski T, Rademacher L, Alva A, Heinecke O, Adkins R, Parkar S, Bhagat R, Lunato J, Beets I, Francis MM, Kowalski JR. Cell non-autonomous signaling through the conserved C. elegans glycoprotein hormone receptor FSHR-1 regulates cholinergic neurotransmission. PLoS Genet 2024; 20:e1011461. [PMID: 39561202 PMCID: PMC11614273 DOI: 10.1371/journal.pgen.1011461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 12/03/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024] Open
Abstract
Modulation of neurotransmission is key for organismal responses to varying physiological contexts such as during infection, injury, or other stresses, as well as in learning and memory and for sensory adaptation. Roles for cell autonomous neuromodulatory mechanisms in these processes have been well described. The importance of cell non-autonomous pathways for inter-tissue signaling, such as gut-to-brain or glia-to-neuron, has emerged more recently, but the cellular mechanisms mediating such regulation remain comparatively unexplored. Glycoproteins and their G protein-coupled receptors (GPCRs) are well-established orchestrators of multi-tissue signaling events that govern diverse physiological processes through both cell-autonomous and cell non-autonomous regulation. Here, we show that follicle stimulating hormone receptor, FSHR-1, the sole Caenorhabditis elegans ortholog of mammalian glycoprotein hormone GPCRs, is important for cell non-autonomous modulation of synaptic transmission. Inhibition of fshr-1 expression reduces muscle contraction and leads to synaptic vesicle accumulation in cholinergic motor neurons. The neuromuscular and locomotor defects in fshr-1 loss-of-function mutants are associated with an underlying accumulation of synaptic vesicles, build-up of the synaptic vesicle priming factor UNC-10/RIM, and decreased synaptic vesicle release from cholinergic motor neurons. Restoration of FSHR-1 to the intestine is sufficient to restore neuromuscular activity and synaptic vesicle localization to fshr-1-deficient animals. Intestine-specific knockdown of FSHR-1 reduces neuromuscular function, indicating FSHR-1 is both necessary and sufficient in the intestine for its neuromuscular effects. Re-expression of FSHR-1 in other sites of endogenous expression, including glial cells and neurons, also restored some neuromuscular deficits, indicating potential cross-tissue regulation from these tissues as well. Genetic interaction studies provide evidence that downstream effectors gsa-1/GαS, acy-1/adenylyl cyclase and sphk-1/sphingosine kinase and glycoprotein hormone subunit orthologs, GPLA-1/GPA2 and GPLB-1/GPB5, are important for intestinal FSHR-1 modulation of the NMJ. Together, our results demonstrate that FSHR-1 modulation directs inter-tissue signaling systems, which promote synaptic vesicle release at neuromuscular synapses.
Collapse
Affiliation(s)
- Morgan Buckley
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - William P. Jacob
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Letitia Bortey
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Makenzi E. McClain
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Alyssa L. Ritter
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Amy Godfrey
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Allyson S. Munneke
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Shankar Ramachandran
- Department of Neurobiology, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, United States of America
| | - Signe Kenis
- Neural Signaling and Circuit Plasticity Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Julie C. Kolnik
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Sarah Olofsson
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Milica Nenadovich
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Tanner Kutoloski
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Lillian Rademacher
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Alexandra Alva
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Olivia Heinecke
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Ryan Adkins
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Shums Parkar
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Reesha Bhagat
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Jaelin Lunato
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| | - Isabel Beets
- Neural Signaling and Circuit Plasticity Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Michael M. Francis
- Department of Neurobiology, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, United States of America
| | - Jennifer R. Kowalski
- Department of Biological Sciences, Butler University, Indianapolis, Indiana, United States of America
| |
Collapse
|
2
|
Buckley M, Jacob WP, Bortey L, McClain M, Ritter AL, Godfrey A, Munneke AS, Ramachandran S, Kenis S, Kolnik JC, Olofsson S, Adkins R, Kutoloski T, Rademacher L, Heinecke O, Alva A, Beets I, Francis MM, Kowalski JR. Cell non-autonomous signaling through the conserved C. elegans glycopeptide hormone receptor FSHR-1 regulates cholinergic neurotransmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.10.578699. [PMID: 38405708 PMCID: PMC10888917 DOI: 10.1101/2024.02.10.578699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Modulation of neurotransmission is key for organismal responses to varying physiological contexts such as during infection, injury, or other stresses, as well as in learning and memory and for sensory adaptation. Roles for cell autonomous neuromodulatory mechanisms in these processes have been well described. The importance of cell non-autonomous pathways for inter-tissue signaling, such as gut-to-brain or glia-to-neuron, has emerged more recently, but the cellular mechanisms mediating such regulation remain comparatively unexplored. Glycoproteins and their G protein-coupled receptors (GPCRs) are well-established orchestrators of multi-tissue signaling events that govern diverse physiological processes through both cell-autonomous and cell non-autonomous regulation. Here, we show that follicle stimulating hormone receptor, FSHR-1, the sole Caenorhabditis elegans ortholog of mammalian glycoprotein hormone GPCRs, is important for cell non-autonomous modulation of synaptic transmission. Inhibition of fshr-1 expression reduces muscle contraction and leads to synaptic vesicle accumulation in cholinergic motor neurons. The neuromuscular and locomotor defects in fshr-1 loss-of-function mutants are associated with an underlying accumulation of synaptic vesicles, build-up of the synaptic vesicle priming factor UNC-10/RIM, and decreased synaptic vesicle release from cholinergic motor neurons. Restoration of FSHR-1 to the intestine is sufficient to restore neuromuscular activity and synaptic vesicle localization to fshr-1- deficient animals. Intestine-specific knockdown of FSHR-1 reduces neuromuscular function, indicating FSHR-1 is both necessary and sufficient in the intestine for its neuromuscular effects. Re-expression of FSHR-1 in other sites of endogenous expression, including glial cells and neurons, also restored some neuromuscular deficits, indicating potential cross-tissue regulation from these tissues as well. Genetic interaction studies provide evidence that downstream effectors gsa-1 / Gα S , acy-1 /adenylyl cyclase and sphk-1/ sphingosine kinase and glycoprotein hormone subunit orthologs, GPLA-1/GPA2 and GPLB-1/GPB5, are important for FSHR-1 modulation of the NMJ. Together, our results demonstrate that FSHR-1 modulation directs inter-tissue signaling systems, which promote synaptic vesicle release at neuromuscular synapses.
Collapse
|
3
|
Baudon A, Clauss Creusot E, Charlet A. [Emergent role of astrocytes in oxytocin-mediated modulatory control of neuronal circuits and brain functions]. Biol Aujourdhui 2023; 216:155-165. [PMID: 36744981 DOI: 10.1051/jbio/2022022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Indexed: 02/07/2023]
Abstract
The neuropeptide oxytocin has been in the focus of scientists for decades due to its profound and pleiotropic effects on physiology, activity of neuronal circuits and behaviors. Until recently, it was believed that oxytocinergic action exclusively occurs through direct activation of neuronal oxytocin receptors. However, several studies demonstrated the existence and functional relevance of astroglial oxytocin receptors in various brain regions in the mouse and rat brain. Astrocytic signaling and activity are critical for many important physiological processes including metabolism, neurotransmitter clearance from the synaptic cleft and integrated brain functions. While it can be speculated that oxytocinergic action on astrocytes predominantly facilitates neuromodulation via the release of gliotransmitters, the precise role of astrocytic oxytocin receptors remains elusive. In this review, we discuss the latest studies on the interaction between the oxytocinergic system and astrocytes, and give details of underlying intracellular cascades.
Collapse
Affiliation(s)
- Angel Baudon
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Etienne Clauss Creusot
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Alexandre Charlet
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| |
Collapse
|
4
|
Baudon A, Clauss Creusot E, Althammer F, Schaaf CP, Charlet A. Emerging role of astrocytes in oxytocin-mediated control of neural circuits and brain functions. Prog Neurobiol 2022; 217:102328. [PMID: 35870680 DOI: 10.1016/j.pneurobio.2022.102328] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/01/2022] [Accepted: 07/18/2022] [Indexed: 11/19/2022]
Abstract
The neuropeptide oxytocin has been in the focus of scientists for decades due to its profound and pleiotropic effects on physiology, activity of neuronal circuits and behaviors, among which sociality. Until recently, it was believed that oxytocinergic action exclusively occurs through direct activation of neuronal oxytocin receptors. However, several studies demonstrated the existence and functional relevance of astroglial oxytocin receptors in various brain regions in the mouse and rat brain. Astrocytic signaling and activity is critical for many important physiological processes including metabolism, neurotransmitter clearance from the synaptic cleft and integrated brain functions. While it can be speculated that oxytocinergic action on astrocytes predominantly facilitates neuromodulation via the release of specific gliotransmitters, the precise role of astrocytic oxytocin receptors remains elusive. In this review, we discuss the latest studies on the interaction between the oxytocinergic system and astrocytes, including detailed information about intracellular cascades, and speculate about future research directions on astrocytic oxytocin signaling.
Collapse
Affiliation(s)
- Angel Baudon
- Centre National de la Recherche Scientifique and University of Strasbourg, Institute of Cellular and Integrative Neuroscience, Strasbourg 67000 France
| | - Etienne Clauss Creusot
- Centre National de la Recherche Scientifique and University of Strasbourg, Institute of Cellular and Integrative Neuroscience, Strasbourg 67000 France
| | | | | | - Alexandre Charlet
- Centre National de la Recherche Scientifique and University of Strasbourg, Institute of Cellular and Integrative Neuroscience, Strasbourg 67000 France.
| |
Collapse
|
5
|
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: 10] [Impact Index Per Article: 3.3] [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
|
6
|
Grassi D, Marraudino M, Garcia-Segura LM, Panzica GC. The hypothalamic paraventricular nucleus as a central hub for the estrogenic modulation of neuroendocrine function and behavior. Front Neuroendocrinol 2022; 65:100974. [PMID: 34995643 DOI: 10.1016/j.yfrne.2021.100974] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022]
Abstract
Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.
Collapse
Affiliation(s)
- D Grassi
- Department of Anatomy, Histology and Neuroscience, Universidad Autonoma de Madrid, Madrid, Spain
| | - M Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - G C Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy; Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy.
| |
Collapse
|
7
|
Domínguez-Ordoñez R, Garcia-Juárez M, Tapia-Hernández S, Luna-Hernández A, Galindo-Madrid ME, Tecamachaltzi-Silvarán MB, Hoffman KL, Pfaus JG, González-Flores O. Oxytocin induces lordosis behavior in female rats through the prostaglandin E2/GnRH signaling system. Horm Behav 2021; 136:105081. [PMID: 34710777 DOI: 10.1016/j.yhbeh.2021.105081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023]
Abstract
Intracerebroventricular (icv) administration of oxytocin (OT) induces robust lordosis behavior (lordosis quotient and lordosis intensity) in estrogen-primed rats. The present study explored the hypothesis that the OT-Prostaglandin E2-GnRH pathway (a pathway produced in astrocytes) is involved in the facilitation of lordosis behavior by icv infusion of OT (2 μg). In Experiment 1, we tested the involvement of the OT receptor (OTR) by infusion of the OTR antagonist, atosiban (ATO). OT-induced lordosis was significantly reduced at both 30 and 120 min by prior infusion of ATO. In Experiment 2, we studied the effects of aspirin (COX2 inhibitor) and ONO-AE3-208 (ONO; EP4 prostaglandin receptor antagonist) on OT-induced lordosis. Infusions of both compounds diminished OT-induced lordosis at both 120 and 240 min. In Experiment 3, the involvement of the GnRH-1 receptor inhibitor antide on OT-induced lordosis was evaluated. Antide significantly inhibited OT-induced lordosis at all times tested. These data indicate that the OT/PGE2/GnRH pathway is involved in the expression of OT-induced lordosis behavior, an effect that may be occurring directly in hypothalamic astrocytes.
Collapse
Affiliation(s)
- Raymundo Domínguez-Ordoñez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | - Marcos Garcia-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | - Sandra Tapia-Hernández
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | - Ailyn Luna-Hernández
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | - Miriam Eli Galindo-Madrid
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | | | - Kurt L Hoffman
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | - James G Pfaus
- Department of Psychology and Life Sciences, Charles University, Prague, Czech Republic; Czech National Institute of Mental Health, Klecany, Czech Republic
| | - Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico.
| |
Collapse
|
8
|
Li D, Liu H, Wang H, Jia S, Wang X, Ling S, Chen G, Liu X, Wang YF. Astrocytic Hydrogen Sulfide Regulates Supraoptic Cellular Activity in the Adaptive Response of Lactating Rats to Chronic Social Stress. ASN Neuro 2021; 13:17590914211043087. [PMID: 34579557 PMCID: PMC8642056 DOI: 10.1177/17590914211043087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Maternal social stress among breastfeeding women can be adapted in chronic process.
However, neuroendocrine mechanisms underlying such adaptation remain to be identified.
Here, we report the effects of 2 hr/day unfamiliar male rat invasion (UMI) stress on
maternal behaviors in lactating rats during postpartum day 8 (UMI8) to postpartum day 12
(UMI12). Rat dams at UMI8 presented signs of maternal anxiety, depression, and attacks
toward male intruder. These changes partially reversed at UMI12 except the sign of
anxiety. In the supraoptic nucleus (SON), UMI12 but not UMI8 significantly increased the
expression of c-Fos and phosphorylated extracellular signal-regulated protein kinase 1/2.
At UMI8 but not UMI12, length of glial fibrillary acidic protein (GFAP, astrocytic
cytoskeletal element) filaments around oxytocin (OT) neurons was significantly longer than
that of their controls; the amount of GFAP fragments at UMI12 was significantly less than
that at UMI8. Expression of cystathionine β-synthase (CBS, enzyme for H2S
synthesis) at UMI12 was significantly higher than that at UMI8. CBS expression did not
change significantly in the somatic zone of the SON but decreased significantly at the
ventral glia lamina at UMI8. In brain slices of the SON, aminooxyacetate (a CBS blocker)
significantly increased the expression of GFAP proteins that were molecularly associated
with CBS. Aminooxyacetate also reduced the firing rate of OT neurons whereas
Na2S, a donor of H2S, increased it. The adaptation during chronic
social stress is possibly attributable to the increased production of H2S by
astrocytes and the subsequent retraction of astrocytic processes around OT neurons.
Collapse
Affiliation(s)
- Dongyang Li
- Department of Physiology, 12455Hainan Medical University, Haikou, China.,Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| | - Haitao Liu
- Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| | - Hongyang Wang
- Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| | - Shuwei Jia
- Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| | - Xiaoran Wang
- Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| | - Shuo Ling
- Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| | - Guichuan Chen
- Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| | - Xiaoyu Liu
- Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, 34707Harbin Medical University, Harbin, China
| |
Collapse
|
9
|
Huang Q, Lian C, Dong Y, Zeng H, Liu B, Xu N, He Z, Guo H. SNAP25 Inhibits Glioma Progression by Regulating Synapse Plasticity via GLS-Mediated Glutaminolysis. Front Oncol 2021; 11:698835. [PMID: 34490096 PMCID: PMC8416623 DOI: 10.3389/fonc.2021.698835] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Background Neuronal activity regulated by synaptic communication exerts an important role in tumorigenesis and progression in brain tumors. Genes for soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) annotated with the function ‘vesicle’ about synaptic connectivity were identified, and synaptosomal-associated protein 25 (SNAP25), one of those proteins, was found to have discrepant expression levels in neuropathies. However, the specific mechanism and prognostic value of SNAP25 during glioma progression remain unclear. Methods Using RNA sequencing data from The Cancer Genome Atlas (TCGA) database, the differential synaptosis-related genes between low grade glioma (LGG) and glioblastoma (GBM) were identified as highly correlated. Cox proportional hazards regression analysis and survival analysis were used to differentiate the outcome of low- and high-risk patients, and the Chinese Glioma Genome Atlas (CGGA) cohort was used for validation of the data set. RT-qPCR, western blot, and immunohistochemistry assays were performed to examine the expression level of SNAP25 in glioma cells and samples. Functional assays were performed to identify the effects of SNAP25 knockdown and overexpression on cell viability, migration, and invasion. Liquid chromatography-high resolution mass spectrometry (LC-MS)-based metabolomics approach was presented for identifying crucial metabolic disturbances in glioma cells. In situ mouse xenograft model was used to investigate the role of SNAP25 in vivo. Then, an immunofluorescence assay of the xenograft tissue was applied to evaluate the expression of the neuronal dendron formation marker-Microtubule Associated Protein 2 (MAP2). Results SNAP25 was decreased in level of expression in glioma tissues and cell lines, and low-level SNAP25 indicated an unfavorable prognosis of glioma patients. SNAP25 inhibited cell proliferation, migration, invasion and fostered glutamine metabolism of glioma cells, exerting a tumor suppressor role. Overexpressed SNAP25 exerted a lower expression level of MAP2, indicating poor neuronal plasticity and connectivity. SNAP25 could regulate glutaminase (GLS)-mediated glutaminolysis, and GLS knockdown could rescue the anti-tumor effect of SNAP25 in glioma cells. Moreover, upregulated SNAP25 also decreased tumor volume and prolonged the overall survival (OS) of the xenograft mouse. Conclusion SNAP25, a tumor suppressor inhibited carcinogenesis of glioma via limiting glutamate metabolism by regulating GLS expression, as well as inhibiting dendritic formation, which could be considered as a novel molecular therapeutic target for glioma.
Collapse
Affiliation(s)
- Qiongzhen Huang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Changlin Lian
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Yaoyuan Dong
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Huijun Zeng
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Boyang Liu
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Ningbo Xu
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Zhenyan He
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Hongbo Guo
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| |
Collapse
|
10
|
Aikins AO, Nguyen DH, Paundralingga O, Farmer GE, Shimoura CG, Brock C, Cunningham JT. Cardiovascular Neuroendocrinology: Emerging Role for Neurohypophyseal Hormones in Pathophysiology. Endocrinology 2021; 162:6247962. [PMID: 33891015 PMCID: PMC8234498 DOI: 10.1210/endocr/bqab082] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Indexed: 11/19/2022]
Abstract
Arginine vasopressin (AVP) and oxytocin (OXY) are released by magnocellular neurosecretory cells that project to the posterior pituitary. While AVP and OXY currently receive more attention for their contributions to affiliative behavior, this mini-review discusses their roles in cardiovascular function broadly defined to include indirect effects that influence cardiovascular function. The traditional view is that neither AVP nor OXY contributes to basal cardiovascular function, although some recent studies suggest that this position might be re-evaluated. More evidence indicates that adaptations and neuroplasticity of AVP and OXY neurons contribute to cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Ato O Aikins
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Dianna H Nguyen
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Obed Paundralingga
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - George E Farmer
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Caroline Gusson Shimoura
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - Courtney Brock
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX 76107, USA
- Correspondence: J. Thomas Cunningham Department of Physiology & Anatomy CBH 338 UNT Health Science Center 3500 Camp Bowie Blvd Fort Worth, TX 76107, USA.
| |
Collapse
|
11
|
Iovino M, Messana T, Tortora A, Giusti C, Lisco G, Giagulli VA, Guastamacchia E, De Pergola G, Triggiani V. Oxytocin Signaling Pathway: From Cell Biology to Clinical Implications. Endocr Metab Immune Disord Drug Targets 2021; 21:91-110. [PMID: 32433011 DOI: 10.2174/1871530320666200520093730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/04/2020] [Accepted: 04/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND In addition to the well-known role played in lactation and parturition, Oxytocin (OT) and OT receptor (OTR) are involved in many other aspects such as the control of maternal and social behavior, the regulation of the growth of the neocortex, the maintenance of blood supply to the cortex, the stimulation of limbic olfactory area to mother-infant recognition bond, and the modulation of the autonomic nervous system via the vagal pathway. Moreover, OT and OTR show antiinflammatory, anti-oxidant, anti-pain, anti-diabetic, anti-dyslipidemic and anti-atherogenic effects. OBJECTIVE The aim of this narrative review is to summarize the main data coming from the literature dealing with the role of OT and OTR in physiology and pathologic conditions focusing on the most relevant aspects. METHODS Appropriate keywords and MeSH terms were identified and searched in Pubmed. Finally, references of original articles and reviews were examined. RESULTS We report the most significant and updated data on the role played by OT and OTR in physiology and different clinical contexts. CONCLUSION Emerging evidence indicates the involvement of OT system in several pathophysiological mechanisms influencing brain anatomy, cognition, language, sense of safety and trust and maternal behavior, with the possible use of exogenous administered OT in the treatment of specific neuropsychiatric conditions. Furthermore, it modulates pancreatic β-cell responsiveness and lipid metabolism leading to possible therapeutic use in diabetic and dyslipidemic patients and for limiting and even reversing atherosclerotic lesions.
Collapse
Affiliation(s)
- Michele Iovino
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Tullio Messana
- Infantile Neuropsychiatry, IRCCS - Institute of Neurological Sciences, Bologna, Italy
| | - Anna Tortora
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Consuelo Giusti
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Giuseppe Lisco
- Hospital Unit of Endocrinology, Perrino Hospital, Brindisi, Italy
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Giovanni De Pergola
- Clinical Nutrition Unit, Medical Oncology, Department of Internal Medicine and Clinical Oncology, University of Bari, School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| |
Collapse
|
12
|
SOX1 Is a Backup Gene for Brain Neurons and Glioma Stem Cell Protection and Proliferation. Mol Neurobiol 2021; 58:2634-2642. [PMID: 33481176 DOI: 10.1007/s12035-020-02240-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022]
Abstract
Failed neuroprotection leads to the initiation of several diseases. SOX1 plays many roles in embryogenesis, oncogenesis, and male sex determination, and can promote glioma stem cell proliferation, invasion, and migration due to its high expression in glioblastoma cells. The functional versatility of the SOX1 gene in malignancy, epilepsy, and Parkinson's disease, as well as its adverse effects on dopaminergic neurons, makes it an interesting research focus. Hence, we collate the most important discoveries relating to the neuroprotective effects of SOX1 in brain cancer and propose hypothesis worthy of SOX1's role in the survival of senescent neuronal cells, its roles in fibroblast cell proliferation, and cell fat for neuroprotection, and the discharge of electrical impulses for homeostasis. Increase in electrical impulses transmitted by senescent cells affects the synthesis of neurotransmitters, which will modify the brain cell metabolism and microenvironment.
Collapse
|
13
|
Liu M, Shen L, Xu M, Wang DQH, Tso P. Estradiol Enhances Anorectic Effect of Apolipoprotein A-IV through ERα-PI3K Pathway in the Nucleus Tractus Solitarius. Genes (Basel) 2020; 11:E1494. [PMID: 33322656 PMCID: PMC7764025 DOI: 10.3390/genes11121494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 01/09/2023] Open
Abstract
Estradiol (E2) enhances the anorectic action of apolipoprotein A-IV (apoA-IV), however, the intracellular mechanisms are largely unclear. Here we reported that the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway was significantly activated by E2 and apoA-IV, respectively, in primary neuronal cells isolated from rat embryonic brainstem. Importantly, the combination of E2 and apoA-IV at their subthreshold doses synergistically activated the PI3K/Akt signaling pathway. These effects, however, were significantly diminished by the pretreatment with LY294002, a selective PI3K inhibitor. E2-induced activation of the PI3K/Akt pathway was through membrane-associated ERα, because the phosphorylation of Akt was significantly increased by PPT, an ERα agonist, and by E2-BSA (E2 conjugated to bovine serum albumin) which activates estrogen receptor on the membrane. Centrally administered apoA-IV at a low dose (0.5 µg) significantly suppressed food intake and increased the phosphorylation of Akt in the nucleus tractus solitarius (NTS) of ovariectomized (OVX) rats treated with E2, but not in OVX rats treated with vehicle. These effects were blunted by pretreatment with LY294002. These results indicate that E2's regulatory role in apoA-IV's anorectic action is through the ERα-PI3K pathway in the NTS. Manipulation of the PI3K/Akt signaling activation in the NTS may provide a novel therapeutic approach for the prevention and the treatment of obesity-related disorders in females.
Collapse
Affiliation(s)
- Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (L.S.); (M.X.); (P.T.)
| | - Ling Shen
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (L.S.); (M.X.); (P.T.)
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (L.S.); (M.X.); (P.T.)
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45237, USA; (L.S.); (M.X.); (P.T.)
| |
Collapse
|
14
|
Early life overnutrition impairs plasticity of non-neuronal brainstem cells and drives obesity in offspring across development in rats. Int J Obes (Lond) 2020; 44:2405-2418. [PMID: 32999409 DOI: 10.1038/s41366-020-00658-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/17/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prevalence of adolescent obesity has increased dramatically, becoming a serious public health concern. While previous evidence suggests that in utero- and early postnatal overnutrition increases adult-onset obesity risk, the neurobiological mechanisms underlying this outcome are not well understood. Non-neuronal cells play an underestimated role in the physiological responses to metabolic/nutrient signals. Hypothalamic glial-mediated inflammation is now considered a contributing factor in the development and perpetuation of obesity; however, attention on the role of gliosis and microglia activation in other nuclei is still needed. METHODS/RESULTS Here, we demonstrate that early life consumption of high-fat/sucrose diet (HFSD) is sufficient to increase offspring body weight, hyperleptinemia and potentially maladaptive cytoarchitectural changes in the brainstem dorsal-vagal-complex (DVC), an essential energy balance processing hub, across postnatal development. Our data demonstrate that pre- and postnatal consumption of HFSD result in increased body weight, hyperleptinemia and dramatically affects the non-neuronal landscape, and therefore the plasticity of the DVC in the developing offspring. CONCLUSIONS Current findings are very provocative, considering the importance of the DVC in appetite regulation, suggesting that HFSD-consumption during early life may contribute to subsequent obesity risk via DVC cytoarchitectural changes.
Collapse
|
15
|
Alvarado JA, Dhande OS, Prosseda PP, Kowal TJ, Ning K, Jabbehdari S, Hu Y, Sun Y. Developmental distribution of primary cilia in the retinofugal visual pathway. J Comp Neurol 2020; 529:1442-1455. [PMID: 32939774 DOI: 10.1002/cne.25029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/21/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022]
Abstract
The mammalian visual system is composed of circuitry connecting sensory input from the retina to the processing core of the visual cortex. The two main retinorecipient brain targets, the superior colliculus (SC) and dorsal lateral geniculate nucleus (dLGN), bridge retinal input and visual output. The primary cilium is a conserved organelle increasingly viewed as a critical sensor for the regulation of developmental and homeostatic pathways in most mammalian cell types. Moreover, cilia have been described as crucial for neurogenesis, neuronal maturation, and survival in the cortex and retina. However, cilia in the visual relay center remain to be fully described. In this study, we characterized the ciliation profile of the SC and dLGN and found that the overall number of ciliated cells declined during development. Interestingly, shorter ciliated cells in both regions were identified as neurons, whose numbers remained stable over time, suggesting that cilia retention is a critical feature for optimal neuronal function in SC and dLGN. Our study suggests that primary cilia are important for neuronal maturation and function in cells of the SC and dLGN.
Collapse
Affiliation(s)
- Jorge A Alvarado
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Onkar S Dhande
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Philipp P Prosseda
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Tia J Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Sayena Jabbehdari
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA.,Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, USA.,Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA.,Palo Alto Veterans Administration, Palo Alto, California, USA
| |
Collapse
|
16
|
Voldsbekk I, Maximov II, Zak N, Roelfs D, Geier O, Due-Tønnessen P, Elvsåshagen T, Strømstad M, Bjørnerud A, Groote I. Evidence for wakefulness-related changes to extracellular space in human brain white matter from diffusion-weighted MRI. Neuroimage 2020; 212:116682. [DOI: 10.1016/j.neuroimage.2020.116682] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/29/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022] Open
|
17
|
Tasker JG, Prager-Khoutorsky M, Teruyama R, Lemos JR, Amstrong WE. Advances in the neurophysiology of magnocellular neuroendocrine cells. J Neuroendocrinol 2020; 32:e12826. [PMID: 31917875 PMCID: PMC7192795 DOI: 10.1111/jne.12826] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Hypothalamic magnocellular neuroendocrine cells have unique electrical properties and a remarkable capacity for morphological and synaptic plasticity. Their large somatic size, their relatively uniform and dense clustering in the supraoptic and paraventricular nuclei, and their large axon terminals in the neurohypophysis make them an attractive target for direct electrophysiological interrogation. Here, we provide a brief review of significant recent findings in the neuroplasticity and neurophysiological properties of these neurones that were presented at the symposium "Electrophysiology of Magnocellular Neurons" during the 13th World Congress on Neurohypophysial Hormones in Ein Gedi, Israel in April 2019. Magnocellular vasopressin (VP) neurones respond directly to hypertonic stimulation with membrane depolarisation, which is triggered by cell shrinkage-induced opening of an N-terminal-truncated variant of transient receptor potential vanilloid type-1 (TRPV1) channels. New findings indicate that this mechanotransduction depends on actin and microtubule cytoskeletal networks, and that direct coupling of the TRPV1 channels to microtubules is responsible for mechanical gating of the channels. Vasopressin neurones also respond to osmostimulation by activation of epithelial Na+ channels (ENaC). It was shown recently that changes in ENaC activity modulate magnocellular neurone basal firing by generating tonic changes in membrane potential. Both oxytocin and VP neurones also undergo robust excitatory synapse plasticity during chronic osmotic stimulation. Recent findings indicate that new glutamate synapses induced during chronic salt loading express highly labile Ca2+ -permeable GluA1 receptors requiring continuous dendritic protein synthesis for synapse maintenance. Finally, recordings from the uniquely tractable neurohypophysial terminals recently revealed an unexpected property of activity-dependent neuropeptide release. A significant fraction of the voltage-dependent neurohypophysial neurosecretion was found to be independent of Ca2+ influx through voltage-gated Ca2+ channels. Together, these findings provide a snapshot of significant new advances in the electrophysiological signalling mechanisms and neuroplasticity of the hypothalamic-neurohypophysial system, a system that continues to make important contributions to the field of neurophysiology.
Collapse
Affiliation(s)
- Jeffrey G. Tasker
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Correspondence: Jeffrey Tasker, PhD, Tulane University, Cell and Molecular Biology Dept, 2000 Percival Stern Hall, New Orleans, LA 70118, USA; .; William Armstrong, PhD, University of Tennessee Health Science Center, Anatomy and Neurobiology Dept and Neuroscience Institute, 855 Monroe Ave, Memphis, TN 38163, USA;
| | | | - Ryoichi Teruyama
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - José R. Lemos
- Department of Microbiology and Physiological Systems & Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA, USA
| | - William E. Amstrong
- Department of Anatomy and Neurobiology and Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
- Correspondence: Jeffrey Tasker, PhD, Tulane University, Cell and Molecular Biology Dept, 2000 Percival Stern Hall, New Orleans, LA 70118, USA; .; William Armstrong, PhD, University of Tennessee Health Science Center, Anatomy and Neurobiology Dept and Neuroscience Institute, 855 Monroe Ave, Memphis, TN 38163, USA;
| |
Collapse
|
18
|
Almeida PG, Nani JV, Oses JP, Brietzke E, Hayashi MA. Neuroinflammation and glial cell activation in mental disorders. Brain Behav Immun Health 2020; 2:100034. [PMID: 38377429 PMCID: PMC8474594 DOI: 10.1016/j.bbih.2019.100034] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 02/07/2023] Open
Abstract
Mental disorders (MDs) are highly prevalent and potentially debilitating complex disorders which causes remain elusive. Looking into deeper aspects of etiology or pathophysiology underlying these diseases would be highly beneficial, as the scarce knowledge in mechanistic and molecular pathways certainly represents an important limitation. Association between MDs and inflammation/neuroinflammation has been widely discussed and accepted by many, as high levels of pro-inflammatory cytokines were reported in patients with several MDs, such as schizophrenia (SCZ), bipolar disorder (BD) and major depression disorder (MDD), among others. Correlation of pro-inflammatory markers with symptoms intensity was also reported. However, the mechanisms underlying the inflammatory dysfunctions observed in MDs are not fully understood yet. In this context, microglial dysfunction has recently emerged as a possible pivotal player, as during the neuroinflammatory response, microglia can be over-activated, and excessive production of pro-inflammatory cytokines, which can modify the kynurenine and glutamate signaling, is reported. Moreover, microglial activation also results in increased astrocyte activity and consequent glutamate release, which are both toxic to the Central Nervous System (CNS). Also, as a result of increased microglial activation in MDs, products of the kynurenine pathway were shown to be changed, influencing then the dopaminergic, serotonergic, and glutamatergic signaling pathways. Therefore, in the present review, we aim to discuss how neuroinflammation impacts on glutamate and kynurenine signaling pathways, and how they can consequently influence the monoaminergic signaling. The consequent association with MDs main symptoms is also discussed. As such, this work aims to contribute to the field by providing insights into these alternative pathways and by shedding light on potential targets that could improve the strategies for pharmacological intervention and/or treatment protocols to combat the main pharmacologically unmatched symptoms of MDs, as the SCZ.
Collapse
Key Words
- AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
- APCs, antigen presenting cells
- BBB, blood-brain barrier
- BD, bipolar disorder
- CCL, C–C motif chemokine ligand
- CLRs, C-type lectin receptors
- CNS, central nervous system
- CSF, cerebrospinal fluid
- CXCL, X–C motif chemokine ligand
- Glia
- IDO, indolamine 2,3-dioxygenase
- IFN, interferon
- IL, interleukin
- IRF, interferon regulatory factor
- Inflammation
- KYNA, kynurenic acid
- MD, mental disorders
- MDD, major depression disorder
- MRI, magnetic resonance imaging
- Mental disorders
- Microglial activation
- NF, necrosis factor
- NMDA, N-methyl-D-aspartate
- NMR, nuclear magnetic resonance
- PPI, prepulse inhibition
- PRRs, pattern recognition receptors
- QUIN, quinolinic acid
- SCZ, schizophrenia
- Schizophrenia
- TGF, tumor growth factor
- TLRs, toll-like receptors
- TNF, tumor necrosis factor
- α7-nAchR, alpha 7 nicotinic acetylcholine receptor
Collapse
Affiliation(s)
- Priscila G.C. Almeida
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - João Victor Nani
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Jean Pierre Oses
- Programa Multicêntrico de Pós-Graduação em Bioquímica e Biologia Molecular, Instituto de Biociências, Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Elisa Brietzke
- Department of Psychiatry, Queen’s University School of Medicine, Kingston, ON, Canada
| | - Mirian A.F. Hayashi
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|
19
|
Kikuchi T, Gonzalez-Soriano J, Kastanauskaite A, Benavides-Piccione R, Merchan-Perez A, DeFelipe J, Blazquez-Llorca L. Volume Electron Microscopy Study of the Relationship Between Synapses and Astrocytes in the Developing Rat Somatosensory Cortex. Cereb Cortex 2020; 30:3800-3819. [PMID: 31989178 PMCID: PMC7233003 DOI: 10.1093/cercor/bhz343] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
In recent years, numerous studies have shown that astrocytes play an important role in neuronal processing of information. One of the most interesting findings is the existence of bidirectional interactions between neurons and astrocytes at synapses, which has given rise to the concept of “tripartite synapses” from a functional point of view. We used focused ion beam milling and scanning electron microscopy (FIB/SEM) to examine in 3D the relationship of synapses with astrocytes that were previously labeled by intracellular injections in the rat somatosensory cortex. We observed that a large number of synapses (32%) had no contact with astrocytic processes. The remaining synapses (68%) were in contact with astrocytic processes, either at the level of the synaptic cleft (44%) or with the pre- and/or post-synaptic elements (24%). Regarding synaptic morphology, larger synapses with more complex shapes were most frequently found within the population that had the synaptic cleft in contact with astrocytic processes. Furthermore, we observed that although synapses were randomly distributed in space, synapses that were free of astrocytic processes tended to form clusters. Overall, at least in the developing rat neocortex, the concept of tripartite synapse only seems to be applicable to a subset of synapses.
Collapse
Affiliation(s)
- Toko Kikuchi
- Center for Biosciences and Informatics, School of Fundamental Science and Technology, Graduate School of Science and Technology, Keio University, 223-8522 Kanagawa, Japan.,Department of Fundamental Neuroscience, University of Lausanne, 1015 Lausanne, Switzerland
| | - Juncal Gonzalez-Soriano
- Departamento de Anatomía, Facultad de Veterinaria, Universidad Complutense, 28040 Madrid, Spain
| | - Asta Kastanauskaite
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Ruth Benavides-Piccione
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain.,Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, 28002 Madrid, Spain
| | - Angel Merchan-Perez
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain.,Departamento de Arquitectura y Tecnología de Sistemas Informáticos, Escuela Técnica Superior de Ingenieros Informáticos, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain.,Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, 28002 Madrid, Spain
| | - Lidia Blazquez-Llorca
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain.,Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain
| |
Collapse
|
20
|
Pathway analysis of glutamate-mediated, calcium-related signaling in glioma progression. Biochem Pharmacol 2020; 176:113814. [PMID: 31954716 DOI: 10.1016/j.bcp.2020.113814] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/13/2020] [Indexed: 12/18/2022]
Abstract
Brain tumors, particularly high-grade glioblastomas, are a crucial public health issue due to poor prognosis and an extremely low survival rate. The glioblastoma multiforme (GBM) grows rapidly within its unique microenvironment that is characterized by active neural communications. Therefore, diverse neurotransmitters not only maintain normal brain functions but also influence glioma progression. To fully appreciate the relationship between neurotransmitters and glioma progression, we reviewed potential neurotransmitter contributors in human GBM and the much less aggressive Low-grade glioma (LGG) by combining previously published data from gene-mutation/mRNA sequencing databases together with protein-protein interaction (PPI) network analysis results. The summarized results indicate that glutamatergic and calcium signaling may provide positive feedback to promote glioma formation through 1) metabolic reprogramming and genetic switching to accelerate glioma duplication and progression; 2) upregulation of cytoskeleton proteins and elevation of intracellular Ca2+ levels to increase glutamate release and facilitate formation of synaptic-like connections with surrounding cells in their microenvironment. The upregulated glutamatergic neuronal activities in turn stimulate glioma growth and signaling. Importantly, the enhanced electrical and molecular signals from both neurons and glia propagate out to enable glioma symptoms such as epilepsy and migraine. The elevated intracellular Ca2+ also activates nitric oxide synthase to produce nitric oxide (NO) that can either promote or inhibit tumorigenesis. By analyzing the network effects for complex interaction among neurotransmitters such as glutamate, Ca2+ and NO in brain tumor progression, especially GBM, we identified the glutamatergic signaling as the potential therapeutic targets and suggest manipulation of glutamatergic signaling may be an effective treatment strategy for this aggressive brain cancer.
Collapse
|
21
|
On the existence of mechanoreceptors within the neurovascular unit of the mammalian brain. Brain Struct Funct 2019; 224:2247-2267. [PMID: 31190162 DOI: 10.1007/s00429-019-01863-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/16/2019] [Indexed: 10/26/2022]
Abstract
We describe a set of perivascular interneurons (PINs) with series of fibro-vesicular complexes (FVCs) throughout the gray matter of the adult rabbit and rat brains. PIN-FVCs are ubiquitous throughout the brain vasculature as detected in Golgi-impregnated specimens. Most PINs are small, aspiny cells with short or long (> 1 mm) axons that split and travel along arterial blood vessels. Upon ramification, axons form FVCs around the arising vascular branches; then, paired axons run parallel to the vessel wall until another ramification ensues, and a new FVC is formed. Cytologically, FVCs consist of clusters of perivascular bulbs (PVBs) encircling the precapillary and capillary wall surrounded by end-feet and the extracellular matrix of endothelial cells and pericytes. A PVB contains mitochondria, multivesicular bodies, and granules with a membranous core, similar to Meissner corpuscles and other mechanoreceptors. Some PVBs form asymmetrical, axo-spinous synapses with presumptive adjacent neurons. PINs appear to correspond to the type 1 nNOS-positive neurons whose FVCs co-label with markers of sensory fiber-terminals surrounded by astrocytic end-feet. The PIN is conserved in adult cats and rhesus monkey specimens. The location, ubiquity throughout the vasculature of the mammalian brain, and cytological organization of the PIN-FVCs suggests that it is a sensory receptor intrinsic to the mammalian neurovascular unit that corresponds to an afferent limb of the sensorimotor feed-back mechanism controlling local blood flow.
Collapse
|
22
|
Leng G, Russell JA. The osmoresponsiveness of oxytocin and vasopressin neurones: Mechanisms, allostasis and evolution. J Neuroendocrinol 2019; 31:e12662. [PMID: 30451331 DOI: 10.1111/jne.12662] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/29/2018] [Accepted: 11/15/2018] [Indexed: 12/27/2022]
Abstract
In the rat supraoptic nucleus, every oxytocin cell projects to the posterior pituitary, and is involved both in reflex milk ejection during lactation and in regulating uterine contractions during parturition. All are also osmosensitive, regulating natriuresis. All are also regulated by signals that control appetite, including the neural and hormonal signals that arise from the gut after food intake and from the sites of energy storage. All are also involved in sexual behaviour, anxiety-related behaviours and social behaviours. The challenge is to understand how a single population of neurones can coherently regulate such a diverse set of functions and adapt to changing physiological states. Their multiple functions arise from complex intrinsic properties that confer sensitivity to a wide range of internal and environmental signals. Many of these properties have a distant evolutionary origin in multifunctional, multisensory neurones of Urbilateria, the hypothesised common ancestor of vertebrates, insects and worms. Their properties allow different patterns of oxytocin release into the circulation from their axon terminals in the posterior pituitary into other brain areas from axonal projections, as well as independent release from their dendrites.
Collapse
Affiliation(s)
- Gareth Leng
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - John A Russell
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
23
|
Role of astrocytes, microglia, and tanycytes in brain control of systemic metabolism. Nat Neurosci 2018; 22:7-14. [PMID: 30531847 DOI: 10.1038/s41593-018-0286-y] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 11/06/2018] [Indexed: 12/28/2022]
Abstract
Astrocytes, microglia, and tanycytes play active roles in the regulation of hypothalamic feeding circuits. These non-neuronal cells are crucial in determining the functional interactions of specific neuronal subpopulations involved in the control of metabolism. Recent advances in biology, optics, genetics, and pharmacology have resulted in the emergence of novel and highly sophisticated approaches for studying hypothalamic neuronal-glial networks. Here we summarize the progress in the field and argue that glial-neuronal interactions provide a core hub integrating food-related cues, interoceptive signals, and internal states to adapt a complex set of physiological responses operating on different timescales to finely tune behavior and metabolism according to metabolic status. This expanding knowledge helps to redefine our understanding of the physiology of food intake and energy metabolism.
Collapse
|
24
|
Anbalagan S, Gordon L, Blechman J, Matsuoka RL, Rajamannar P, Wircer E, Biran J, Reuveny A, Leshkowitz D, Stainier DYR, Levkowitz G. Pituicyte Cues Regulate the Development of Permeable Neuro-Vascular Interfaces. Dev Cell 2018; 47:711-726.e5. [PMID: 30449506 DOI: 10.1016/j.devcel.2018.10.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 09/07/2018] [Accepted: 10/15/2018] [Indexed: 11/30/2022]
Abstract
The hypothalamo-neurohypophyseal system (HNS) regulates homeostasis through the passage of neurohormones and blood-borne proteins via permeable blood capillaries that lack the blood-brain barrier (BBB). Why neurohypophyseal capillaries become permeable while the neighboring vasculature of the brain forms BBB remains unclear. We show that pituicytes, the resident astroglial cells of the neurohypophysis, express genes that are associated with BBB breakdown during neuroinflammation. Pituicyte-enriched factors provide a local microenvironment that instructs a permeable neurovascular conduit. Thus, genetic and pharmacological perturbations of Vegfa and Tgfβ3 affected HNS vascular morphogenesis and permeability and impaired the expression of the fenestral marker plvap. The anti-inflammatory agent dexamethasone decreased HNS permeability and downregulated the pituicyte-specific cyp26b gene, encoding a retinoic acid catabolic enzyme. Inhibition of Cyp26b activity led to upregulation of tight junction protein Claudin-5 and decreased permeability. We conclude that pituicyte-derived factors regulate the "decision" of endothelial cells to adopt a permeable endothelial fate instead of forming a BBB.
Collapse
Affiliation(s)
- Savani Anbalagan
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Ludmila Gordon
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Ryota L Matsuoka
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Preethi Rajamannar
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Einav Wircer
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Jakob Biran
- Department of Poultry and Aquaculture, Agricultural Research Organization, Rishon Letziyon 7528809, Israel
| | - Adriana Reuveny
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Dena Leshkowitz
- Bioinformatics Unit, LSCF Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel.
| |
Collapse
|
25
|
Up to Date on Astrocytes. Int Neurourol J 2018; 22:S104-105. [PMID: 30396258 PMCID: PMC6234723 DOI: 10.5213/inj.1820edi.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
26
|
Prepubertal Development of GABAergic Transmission to Gonadotropin-Releasing Hormone (GnRH) Neurons and Postsynaptic Response Are Altered by Prenatal Androgenization. J Neurosci 2018; 38:2283-2293. [PMID: 29374136 DOI: 10.1523/jneurosci.2304-17.2018] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/29/2017] [Accepted: 01/20/2018] [Indexed: 11/21/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons regulate reproduction through pulsatile GnRH release. Women with polycystic ovary syndrome (PCOS) have persistently elevated luteinizing hormone release frequency, reflecting GnRH release; this exacerbates hyperandrogenemia and disrupted reproductive cycles that are characteristic of this disorder. Clinical evidence suggests that neuroendocrine features of PCOS may manifest peripubertally. Adult mice prenatally exposed to androgens (PNA) mimic several reproductive features of PCOS. GnRH neurons from these mice have increased firing activity and receive increased GABAergic transmission, which is excitatory. When changes emerge during development is unknown. To study the typical postnatal development of GABAergic transmission and the effects of PNA treatment and sex, whole-cell voltage-clamp recordings were made of GABAergic postsynaptic currents (PSCs) in GnRH neurons in brain slices from prepubertal through adult control and PNA female and male mice. GABAergic transmission was present by 1 week of age in females and males and increased in frequency, reaching adult levels at 3 and 4 weeks, respectively. GABAergic PSC frequency was elevated in 3-week-old PNA versus control females. PSC frequency in both controls and PNA mice was activity independent, suggesting that PNA induces changes in synapse organization. PNA also alters the functional response of GnRH neurons to GABA. GABA induced firing in fewer neurons from 3-week-old PNA than control females; membrane potential depolarization induced by GABA was also reduced in cells from PNA mice at this age. PNA thus induces changes during development in the presynaptic organization of the GABAergic network afferent to GnRH neurons as well as the postsynaptic GnRH neuron response, both of which may contribute to adult reproductive dysfunction.SIGNIFICANCE STATEMENT The central neuronal network that regulates reproduction is overactive in polycystic ovary syndrome (PCOS), a leading cause of infertility. Recent evidence of neuroendocrine dysfunction in midpubertal girls suggests that the pathophysiological mechanisms underlying PCOS may arise before pubertal maturation. Prenatal exposure to androgens (PNA) in mice mimics several neuroendocrine features of PCOS. GABAergic transmission to gonadotropin-releasing hormone (GnRH) neurons is important for reproduction and is increased in adult PNA mice. The typical development of this network and when changes with PNA and sex arise relative to puberty are unknown. These studies provide evidence that PNA alters prepubertal development of the GABAergic network afferent to GnRH neurons, including both the presynaptic organization and postsynaptic response. These changes may contribute to reproductive dysfunction in adults.
Collapse
|
27
|
Augustine RA, Seymour AJ, Campbell RE, Grattan DR, Brown CH. Integrative neuro-humoral regulation of oxytocin neuron activity in pregnancy and lactation. J Neuroendocrinol 2018; 30. [PMID: 29323764 DOI: 10.1111/jne.12569] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/07/2018] [Indexed: 02/02/2023]
Abstract
Oxytocin is required for normal birth and lactation. Oxytocin is synthesised by hypothalamic supraoptic and paraventricular nuclei neurons and is released into the circulation from the posterior pituitary gland. Under basal conditions, circulating oxytocin levels are relatively constant but during birth and lactation, pulsatile oxytocin release triggers rhythmic contraction of the uterus during birth and milk ejection during suckling. Oxytocin levels are principally determined by the pattern of action potential firing that is, in turn, determined by the interplay between the intrinsic properties of the oxytocin neurons, regulation of their excitability by surrounding glia as well as by synaptic drive from their afferent inputs. During birth and suckling, oxytocin neurons fire high-frequency bursts of action potentials that are coordinated across the population of neurons and these bursts underpin the pulsatile secretion of oxytocin required for normal birth and lactation. Neuroglial regulation of oxytocin neurons changes during pregnancy to favour burst firing. However, these changes still require afferent input activity to drive activity. While it has long been known that noradrenergic inputs to oxytocin neurons are activated during birth and lactation, the involvement of other afferent inputs is less clear. Here, we provide a brief overview of the current understanding of the mechanisms that regulate oxytocin neuron activity during pregnancy and lactation, and focus on recent evidence from our laboratory identifying an input that increases kisspeptin production to excite oxytocin neurons in late pregnancy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Rachael A Augustine
- Department of Physiology Brain Health Research Centre, Centre for Neuroendocrinology
| | - Alexander J Seymour
- Department of Physiology Brain Health Research Centre, Centre for Neuroendocrinology
| | - Rebecca E Campbell
- Department of Physiology Brain Health Research Centre, Centre for Neuroendocrinology
| | - David R Grattan
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Colin H Brown
- Department of Physiology Brain Health Research Centre, Centre for Neuroendocrinology
| |
Collapse
|
28
|
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: 83] [Impact Index Per Article: 11.9] [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
|
29
|
Canine dorsal root ganglia satellite glial cells represent an exceptional cell population with astrocytic and oligodendrocytic properties. Sci Rep 2017; 7:13915. [PMID: 29066783 PMCID: PMC5654978 DOI: 10.1038/s41598-017-14246-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/06/2017] [Indexed: 12/21/2022] Open
Abstract
Dogs can be used as a translational animal model to close the gap between basic discoveries in rodents and clinical trials in humans. The present study compared the species-specific properties of satellite glial cells (SGCs) of canine and murine dorsal root ganglia (DRG) in situ and in vitro using light microscopy, electron microscopy, and immunostainings. The in situ expression of CNPase, GFAP, and glutamine synthetase (GS) has also been investigated in simian SGCs. In situ, most canine SGCs (>80%) expressed the neural progenitor cell markers nestin and Sox2. CNPase and GFAP were found in most canine and simian but not murine SGCs. GS was detected in 94% of simian and 71% of murine SGCs, whereas only 44% of canine SGCs expressed GS. In vitro, most canine (>84%) and murine (>96%) SGCs expressed CNPase, whereas GFAP expression was differentially affected by culture conditions and varied between 10% and 40%. However, GFAP expression was induced by bone morphogenetic protein 4 in SGCs of both species. Interestingly, canine SGCs also stimulated neurite formation of DRG neurons. These findings indicate that SGCs represent an exceptional, intermediate glial cell population with phenotypical characteristics of oligodendrocytes and astrocytes and might possess intrinsic regenerative capabilities in vivo.
Collapse
|
30
|
Zhang M, Biancardi VC, Stern JE. An increased extrasynaptic NMDA tone inhibits A-type K + current and increases excitability of hypothalamic neurosecretory neurons in hypertensive rats. J Physiol 2017; 595:4647-4661. [PMID: 28378360 PMCID: PMC5509869 DOI: 10.1113/jp274327] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/31/2017] [Indexed: 12/27/2022] Open
Abstract
KEY POINTS A functional coupling between extrasynaptic NMDA receptors (eNMDARs) and the A-type K+ current (IA ) influences homeostatic firing responses of magnocellular neurosecretory cells (MNCs) to a physiological challenge. However, whether an altered eNMDAR-IA coupling also contributes to exacerbated MNC activity and neurohumoral activation during disease states is unknown. We show that activation of eNMDARs by exogenously applied NMDA inhibited IA in MNCs obtained from sham, but not in MNCs from renovascular hypertensive (RVH) rats. Neither the magnitude of the exogenously evoked NMDA current nor the expression of NMDAR subunits were altered in RVH rats. Conversely, we found that a larger endogenous glutamate tone, which was not due to blunted glutamate transport activity, led to the sustained activation of eNMDARs that tonically inhibited IA , contributing in turn to higher firing activity in RVH rats. Our studies show that exacerbated activation of eNMDARs by endogenous glutamate contributes to tonic inhibition of IA and enhanced MNC excitability in RVH rats. ABSTRACT We recently showed that a functional coupling between extrasynaptic NMDA receptors (eNMDARs) and the A-type K+ current (IA ) influences the firing activity of hypothalamic magnocellular neurosecretory neurons (MNCs), as well as homeostatic adaptive responses to a physiological challenge. Here, we aimed to determine whether changes in the eNMDAR-IA coupling also contributed to exacerbated MNC activity during disease states. We used a combination of patch-clamp electrophysiology and real-time PCR in MNCs in sham and renovascular hypertensive (RVH) rats. Activation of eNMDARs by exogenously applied NMDA inhibited IA in sham rats, but this effect was largely blunted in RVH rats. The blunted response was not due to changes in eNMDAR expression and/or function, since neither NMDA current magnitude or reversal potential, nor the levels of NR1-NR2A-D subunit expression were altered in RVH rats. Conversely, we found a larger endogenous glutamate tone, resulting in the sustained activation of eNMDARs that tonically inhibited IA and contributed also to higher ongoing firing activity in RVH rats. The enhanced endogenous glutamate tone in RVH rats was not due to blunted glutamate transporter activity. Rather, a higher transporter activity was observed, which possibly acted as a compensatory mechanism in the face of the elevated endogenous tone. In summary, our studies indicate that an elevated endogenous glutamate tone results in an exacerbated activation of eNMDARs, which in turn contributes to diminished IA magnitude and increased firing activity of MNCs from hypertensive rats.
Collapse
Affiliation(s)
- Meng Zhang
- Department of PhysiologyMedical College of GeorgiaAugusta University1120 15th StreetAugustaGA30912USA
| | - Vinicia C. Biancardi
- Department of PhysiologyMedical College of GeorgiaAugusta University1120 15th StreetAugustaGA30912USA
| | - Javier E. Stern
- Department of PhysiologyMedical College of GeorgiaAugusta University1120 15th StreetAugustaGA30912USA
| |
Collapse
|
31
|
Agostinelli LJ, Ferrari LL, Mahoney CE, Mochizuki T, Lowell BB, Arrigoni E, Scammell TE. Descending projections from the basal forebrain to the orexin neurons in mice. J Comp Neurol 2017; 525:1668-1684. [PMID: 27997037 PMCID: PMC5806522 DOI: 10.1002/cne.24158] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 12/23/2022]
Abstract
The orexin (hypocretin) neurons play an essential role in promoting arousal, and loss of the orexin neurons results in narcolepsy, a condition characterized by chronic sleepiness and cataplexy. The orexin neurons excite wake-promoting neurons in the basal forebrain (BF), and a reciprocal projection from the BF back to the orexin neurons may help promote arousal and motivation. The BF contains at least three different cell types (cholinergic, glutamatergic, and γ-aminobutyric acid (GABA)ergic neurons) across its different regions (medial septum, diagonal band, magnocellular preoptic area, and substantia innominata). Given the neurochemical and anatomical heterogeneity of the BF, we mapped the pattern of BF projections to the orexin neurons across multiple BF regions and neuronal types. We performed conditional anterograde tracing using mice that express Cre recombinase only in neurons producing acetylcholine, glutamate, or GABA. We found that the orexin neurons are heavily apposed by axon terminals of glutamatergic and GABAergic neurons of the substantia innominata (SI) and magnocellular preoptic area, but there was no innervation by the cholinergic neurons. Channelrhodopsin-assisted circuit mapping (CRACM) demonstrated that glutamatergic SI neurons frequently form functional synapses with the orexin neurons, but, surprisingly, functional synapses from SI GABAergic neurons were rare. Considering their strong reciprocal connections, BF and orexin neurons likely work in concert to promote arousal, motivation, and other behaviors. J. Comp. Neurol. 525:1668-1684, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lindsay J Agostinelli
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Loris L Ferrari
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Carrie E Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Takatoshi Mochizuki
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Bradford B Lowell
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Miyata S. Advances in Understanding of Structural Reorganization in the Hypothalamic Neurosecretory System. Front Endocrinol (Lausanne) 2017; 8:275. [PMID: 29089925 PMCID: PMC5650978 DOI: 10.3389/fendo.2017.00275] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/28/2017] [Indexed: 12/18/2022] Open
Abstract
The hypothalamic neurosecretory system synthesizes neuropeptides in hypothalamic nuclei and releases them from axonal terminals into the circulation in the neurohypophysis (NH) and median eminence (ME). This system plays a crucial role in regulating body fluid homeostasis and social behaviors as well as reproduction, growth, metabolism, and stress responses, and activity-dependent structural reorganization has been reported. Current knowledge on dynamic structural reorganization in the NH and ME, in which the axonal terminals of neurosecretory neurons directly contact the basement membrane (BM) of a fenestrated vasculature, is discussed herein. Glial cells, pituicytes in the NH and tanycytes in the ME, engulf axonal terminals and interpose their cellular processes between axonal terminals and the BM when hormonal demands are low. Increasing demands for neurosecretion result in the retraction of the cellular processes of glial cells from axonal terminals and the BM, permitting increased neurovascular contact. The shape conversion of pituicytes and tanycytes is mediated by neurotransmitters and sex steroid hormones, respectively. The NH and ME have a rough vascular BM profile of wide perivascular spaces and specialized extension structures called "perivascular protrusions." Perivascular protrusions, the insides of which are occupied by the cellular processes of vascular mural cells pericytes, contribute to increasing neurovascular contact and, thus, the efficient diffusion of hypothalamic neuropeptides. A chronic physiological stimulation has been shown to increase perivascular protrusions via the shape conversion of pericytes and the profile of the vascular surface. Continuous angiogenesis occurs in the NH and ME of healthy normal adult rodents depending on the signaling of vascular endothelial growth factor (VEGF). The inhibition of VEGF signaling suppresses the proliferation of endothelial cells (ECs) and promotes their apoptosis, which results in decreases in the population of ECs and axonal terminals. Pituicytes and tanycytes are continuously replaced by the proliferation and differentiation of stem/progenitor cells, which may be regulated by matching those of ECs and axonal terminals. In conclusion, structural reorganization in the NH and ME is caused by the activity-dependent shape conversion of glial cells and vascular mural cells as well as the proliferation of endothelial and glial cells by angiogenesis and gliogenesis, respectively.
Collapse
Affiliation(s)
- Seiji Miyata
- Department of Applied Biology, The Center for Advanced Insect Research Promotion (CAIRP), Kyoto Institute of Technology, Kyoto, Japan
- *Correspondence: Seiji Miyata,
| |
Collapse
|
33
|
Sun H, Li R, Xu S, Liu Z, Ma X. Hypothalamic Astrocytes Respond to Gastric Mucosal Damage Induced by Restraint Water-Immersion Stress in Rat. Front Behav Neurosci 2016; 10:210. [PMID: 27847472 PMCID: PMC5088369 DOI: 10.3389/fnbeh.2016.00210] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/17/2016] [Indexed: 12/20/2022] Open
Abstract
Restraint water-immersion stress (RWIS), a compound stress model, includes both psychological and physical stimulation. Studies have shown that neurons in the hypothalamus are involved in RWIS, but the role of astrocytes and the interactions between astrocytes and neurons in RWIS are not clear. Here, we tested our hypothesis that hypothalamus astrocytes are involved in RWIS and interact with neurons to regulate gastric mucosal damage induced by RWIS. The expression of Glial fibrillary acidic protein (GFAP) and c-Fos in the paraventricular nucleus (PVN) and supraoptic nucleus (SON) significantly increased following the RWIS. GFAP and c-Fos expression are similar in the temporal pattern, peaked at 1 h after the RWIS, then reduced gradually, and reached a maximal level again at 5 h which show “double-peak” characteristics. Intracerebroventricular administration of astroglial toxin L-a-aminoadipate (L-AA) and c-Fos antisense oligodeoxy nucleotides (ASO) both decreased RWIS-induced gastric mucosal damage. Results of immunohistochemistry assay revealed that both L-AA and ASO decreased the activation of astrocytes and neurons in the hypothalamus by RWIS. These results showed that hypothalamus neuron-astrocyte “network” involved in gastric mucosal damage induced by RWIS. This study may offer theoretical basis for some novel therapeutic strategies for RWIS-induced gastric ulcers.
Collapse
Affiliation(s)
- Haiji Sun
- College of Life Science, Shandong Normal University Jinan, China
| | - Ruisheng Li
- Research Center for Clinical and Translational Medicine, 302 Hospital of PLA Beijing, China
| | - Shiguo Xu
- College of Life Science, Shandong Normal University Jinan, China
| | - Zhen Liu
- College of Life Science, Shandong Normal University Jinan, China
| | - Xiaoli Ma
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University Jinan, China
| |
Collapse
|
34
|
Lozić M, Tasić T, Martin A, Greenwood M, Šarenac O, Hindmarch C, Paton JF, Murphy D, Japundžić-Žigon N. Over-expression of V1A receptors in PVN modulates autonomic cardiovascular control. Pharmacol Res 2016; 114:185-195. [PMID: 27810519 DOI: 10.1016/j.phrs.2016.10.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/07/2016] [Accepted: 10/28/2016] [Indexed: 11/29/2022]
Abstract
The hypothalamic paraventricular nucleus (PVN) is a key integrative site for the neuroendocrine control of the circulation and of the stress response. It is also a major source of the neuropeptide hormone vasopressin (VP), and co-expresses V1a receptors (V1aR). We thus sought to investigate the role of V1aR in PVN in cardiovascular control in response to stress. Experiments were performed in male Wistar rats equipped with radiotelemetric device. The right PVN was transfected with adenoviral vectors (Ads) engineered to over-express V1aR along with an enhanced green fluorescent protein (eGFP) tag. Control groups were PVN transfected with Ads expressing eGFP alone, or wild-type rats (Wt). Rats were recorded with and without selective blockade of V1aR (V1aRX) in PVN under both baseline and stressed conditions. Blood pressure (BP), heart rate (HR), their short-term variabilities, and baroreflex sensitivity (BRS) were evaluated using spectral analysis and the sequence method, respectively. Under baseline physiological conditions,V1aR rats exhibited reduced BRS and a marked increase of BP and HR variability during exposure to stress. These effects were all prevented by V1aRX pretreatment. In Wt rats, V1aRX did not modify cardiovascular parameters under baseline conditions, and prevented BP variability increase by stress. However, V1aRX pretreatment did not modify baroreflex desensitization by stress in either rat strain. It follows that increased expression of V1aR in PVN influences autonomic cardiovascular regulation and demarcates vulnerability to stress. We thus suggest a possible role of hypothalamic V1aR in cardiovascular pathology.
Collapse
Affiliation(s)
- Maja Lozić
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine University of Belgrade, 11000 Belgrade, Serbia
| | - Tatjana Tasić
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine University of Belgrade, 11000 Belgrade, Serbia
| | - Andrew Martin
- Molecular Neuroendocrinology Research Group, The Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, England BS1 3NY, United Kingdom
| | - Michael Greenwood
- Molecular Neuroendocrinology Research Group, The Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, England BS1 3NY, United Kingdom
| | - Olivera Šarenac
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine University of Belgrade, 11000 Belgrade, Serbia; Molecular Neuroendocrinology Research Group, The Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, England BS1 3NY, United Kingdom
| | - Charles Hindmarch
- Molecular Neuroendocrinology Research Group, The Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, England BS1 3NY, United Kingdom; Department of Physiology, University of Malaya, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Julian F Paton
- School of Physiology and Pharmacology, University of Bristol, Bristol, England BS8 1TD, United Kingdom
| | - David Murphy
- Molecular Neuroendocrinology Research Group, The Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, England BS1 3NY, United Kingdom; Department of Physiology, University of Malaya, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Nina Japundžić-Žigon
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine University of Belgrade, 11000 Belgrade, Serbia.
| |
Collapse
|
35
|
Boury-Jamot B, Halfon O, Magistretti PJ, Boutrel B. Lactate release from astrocytes to neurons contributes to cocaine memory formation. Bioessays 2016; 38:1266-1273. [DOI: 10.1002/bies.201600118] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Benjamin Boury-Jamot
- Department of Psychiatry; Centre for Psychiatric Neuroscience; Lausanne University Hospital; Lausanne Switzerland
- Brain Mind Institute; Ecole Polytechnique Fédérale de Lausanne (EPFL); Lausanne Switzerland
| | - Olivier Halfon
- Division of Child and Adolescent Psychiatry; Department of Psychiatry; Lausanne University Hospital; Lausanne Switzerland
| | - Pierre J. Magistretti
- Department of Psychiatry; Centre for Psychiatric Neuroscience; Lausanne University Hospital; Lausanne Switzerland
- Brain Mind Institute; Ecole Polytechnique Fédérale de Lausanne (EPFL); Lausanne Switzerland
- King Abdullah University of Science and Technology (KAUST); Thuwal Saudi Arabia
| | - Benjamin Boutrel
- Department of Psychiatry; Centre for Psychiatric Neuroscience; Lausanne University Hospital; Lausanne Switzerland
- Division of Child and Adolescent Psychiatry; Department of Psychiatry; Lausanne University Hospital; Lausanne Switzerland
| |
Collapse
|
36
|
Stern JE, Son S, Biancardi VC, Zheng H, Sharma N, Patel KP. Astrocytes Contribute to Angiotensin II Stimulation of Hypothalamic Neuronal Activity and Sympathetic Outflow. Hypertension 2016; 68:1483-1493. [PMID: 27698069 DOI: 10.1161/hypertensionaha.116.07747] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/17/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023]
Abstract
Angiotensin II (AngII) is a key neuropeptide that acting within the brain hypothalamic paraventricular nucleus regulates neurohumoral outflow to the circulation. Moreover, an exacerbated AngII action within the paraventricular nucleus contributes to neurohumoral activation in hypertension. Although AngII effects involve changes in paraventricular nucleus neuronal activity, the precise underlying mechanisms, cellular targets, and distribution of AngII receptors within the paraventricular nucleus remain largely unknown. Thus, whether AngII effects involve direct actions on paraventricular neurons, or whether it acts via intermediary cells, such as astrocytes, is still controversial. To address this important gap in our knowledge, we used a multidisciplinary approach combining patch-clamp electrophysiology in presympathetic paraventricular neurons and astrocytes, along with in vivo sympathetic nerve recordings and astrocyte-targeted gene manipulations. We present evidence for a novel mechanism underlying central AngII actions, which involves astrocytes as major intermediary cellular targets. We found that AngII type 1 receptor mRNA is expressed in paraventricular astrocytes. Moreover, we report that AngII inhibited glutamate transporter function, increasing in turn extracellular glutamate levels. This resulted in the activation of neuronal extrasynaptic NMDA (N-methyl-d-aspartate) receptors, increased presympathetic neuronal activity, enhanced sympathoexcitatory outflow, and increased blood pressure. Together, our studies support astrocytes as critical intermediary cell types mediating brain AngII regulation of the circulation and indicate that AngII-mediated neuronal and sympathoexcitatory effects are dependent on a unique neuroglial signaling modality involving nonsynaptic glutamate transmission.
Collapse
Affiliation(s)
- Javier E Stern
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.).
| | - Sookjin Son
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| | - Vinicia C Biancardi
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| | - Hong Zheng
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| | - Neeru Sharma
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| | - Kaushik P Patel
- From the Department of Physiology, Augusta University, GA (J.E.S., S.S., V.C.B.); and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (H.Z., N.S., K.P.P.)
| |
Collapse
|
37
|
Abstract
The posterior pituitary gland secretes oxytocin and vasopressin (the antidiuretic hormone) into the blood system. Oxytocin is required for normal delivery of the young and for delivery of milk to the young during lactation. Vasopressin increases water reabsorption in the kidney to maintain body fluid balance and causes vasoconstriction to increase blood pressure. Oxytocin and vasopressin secretion occurs from the axon terminals of magnocellular neurons whose cell bodies are principally found in the hypothalamic supraoptic nucleus and paraventricular nucleus. The physiological functions of oxytocin and vasopressin depend on their secretion, which is principally determined by the pattern of action potentials initiated at the cell bodies. Appropriate secretion of oxytocin and vasopressin to meet the challenges of changing physiological conditions relies mainly on integration of afferent information on reproductive, osmotic, and cardiovascular status with local regulation of magnocellular neurons by glia as well as intrinsic regulation by the magnocellular neurons themselves. This review focuses on the control of magnocellular neuron activity with a particular emphasis on their regulation by reproductive function, body fluid balance, and cardiovascular status. © 2016 American Physiological Society. Compr Physiol 6:1701-1741, 2016.
Collapse
Affiliation(s)
- Colin H Brown
- Brain Health Research Centre, Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
38
|
Vargová L, Syková E. Astrocytes and extracellular matrix in extrasynaptic volume transmission. Philos Trans R Soc Lond B Biol Sci 2015; 369:20130608. [PMID: 25225101 DOI: 10.1098/rstb.2013.0608] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Volume transmission is a form of intercellular communication that does not require synapses; it is based on the diffusion of neuroactive substances across the brain extracellular space (ECS) and their binding to extrasynaptic high-affinity receptors on neurons or glia. Extracellular diffusion is restricted by the limited volume of the ECS, which is described by the ECS volume fraction α, and the presence of diffusion barriers, reflected by tortuosity λ, that are created, for example, by fine astrocytic processes or extracellular matrix (ECM) molecules. Organized astrocytic processes, ECM scaffolds or myelin sheets channel the extracellular diffusion so that it is facilitated in a certain direction, i.e. anisotropic. The diffusion properties of the ECS are profoundly influenced by various processes such as the swelling and morphological rebuilding of astrocytes during either transient or persisting physiological or pathological states, or the remodelling of the ECM in tumorous or epileptogenic tissue, during Alzheimer's disease, after enzymatic treatment or in transgenic animals. The changing diffusion properties of the ECM influence neuron-glia interaction, learning abilities, the extent of neuronal damage and even cell migration. From a clinical point of view, diffusion parameter changes occurring during pathological states could be important for diagnosis, drug delivery and treatment.
Collapse
Affiliation(s)
- Lýdia Vargová
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic Department of Neuroscience, Institute of Experimental Medicine AS CR, Prague, Czech Republic
| | - Eva Syková
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic Department of Neuroscience, Institute of Experimental Medicine AS CR, Prague, Czech Republic
| |
Collapse
|
39
|
Lee KM, Chiu KB, Didier PJ, Baker KC, MacLean AG. Naltrexone treatment reverses astrocyte atrophy and immune dysfunction in self-harming macaques. Brain Behav Immun 2015; 50:288-297. [PMID: 26191654 PMCID: PMC4631668 DOI: 10.1016/j.bbi.2015.07.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/30/2015] [Accepted: 07/16/2015] [Indexed: 01/19/2023] Open
Abstract
The role of glia in the development and treatment of behavioral abnormalities is understudied. Recent reports have observed glial activation in several disorders, including depression, autism spectrum disorders and self-injurious behaviors (SIB). In the current study, we examined SIB in the physiologically and anatomically relevant nonhuman primate (NHP) model. At the Tulane National Primate Research Center (TNPRC), approximately 5% of singly housed macaques develop symptoms of SIB. We have previously demonstrated that naltrexone hydrochloride can be effective in reducing SIB. We have also demonstrated that the astrocytes of animals with SIB are distinctly atrophic and display heightened innate immune activation compared with control animals. We have added a third group of animals (five macaques identified with SIB and treated with oral naltrexone at a dose of 3.2mg/kg) to the previous cohort (six macaques with a history of SIB but not treated, and nine animals with no history of SIB) for this study. Gray and white matter astrocytes from frontal cortical tissue were examined following necropsy. Innate immune activation of astrocytes, which was increased in SIB animals, was markedly decreased in animals receiving naltrexone, as was atrophy of both grey and white matter astrocytes. This was concomitant with improved behavioral correlates. Preventing astrocyte activation in select areas of the brain to reduce injurious behavior is an innovative concept with implications for mental health studies. Differences in multiple areas of primate brain would help determine how self-injurious behavior develops. These studies suggest a stronger role for astrocytes in the cellular events associated with self-injurious behaviors.
Collapse
Affiliation(s)
- Kim M. Lee
- Tulane National Primate Research Center, Covington, LA, USA,Tulane Program in Biomedical Science, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kevin B. Chiu
- Tulane National Primate Research Center, Covington, LA, USA,Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | | | - Kate C. Baker
- Tulane National Primate Research Center, Covington, LA, USA,Department of Psychology, Tulane University, New Orleans, LA, USA,Tulane Program in Neuroscience, Tulane University, New Orleans, LA, USA
| | - Andrew G. MacLean
- Tulane National Primate Research Center, Covington, LA, USA,Tulane Program in Biomedical Science, Tulane University School of Medicine, New Orleans, LA, USA,Tulane Program in Neuroscience, Tulane University, New Orleans, LA, USA,Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA,Corresponding author: Andrew G. MacLean, Tulane National Primate Research Center, Covington, LA, 70433. ‘phone: 985-871-6489
| |
Collapse
|
40
|
Abstract
Prolactin (PRL) released from lactotrophs of the anterior pituitary gland in response to the suckling by the offspring is the major hormonal signal responsible for stimulation of milk synthesis in the mammary glands. PRL secretion is under chronic inhibition exerted by dopamine (DA), which is released from neurons of the arcuate nucleus of the hypothalamus into the hypophyseal portal vasculature. Suckling by the young activates ascending systems that decrease the release of DA from this system, resulting in enhanced responsiveness to one or more PRL-releasing hormones, such as thyrotropin-releasing hormone. The neuropeptide oxytocin (OT), synthesized in magnocellular neurons of the hypothalamic supraoptic, paraventricular, and several accessory nuclei, is responsible for contracting the myoepithelial cells of the mammary gland to produce milk ejection. Electrophysiological recordings demonstrate that shortly before each milk ejection, the entire neurosecretory OT population fires a synchronized burst of action potentials (the milk ejection burst), resulting in release of OT from nerve terminals in the neurohypophysis. Both of these neuroendocrine systems undergo alterations in late gestation that prepare them for the secretory demands of lactation, and that reduce their responsiveness to stimuli other than suckling, especially physical stressors. The demands of milk synthesis and release produce a condition of negative energy balance in the suckled mother, and, in laboratory rodents, are accompanied by a dramatic hyperphagia. The reduction in secretion of the adipocyte hormone, leptin, a hallmark of negative energy balance, may be an important endocrine signal to hypothalamic systems that integrate lactation-associated food intake with neuroendocrine systems.
Collapse
Affiliation(s)
- William R Crowley
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah Health Sciences Center, Salt Lake City, Utah
| |
Collapse
|
41
|
Bosler O, Girardet C, Franc JL, Becquet D, François-Bellan AM. Structural plasticity of the circadian timing system. An overview from flies to mammals. Front Neuroendocrinol 2015; 38:50-64. [PMID: 25703789 DOI: 10.1016/j.yfrne.2015.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/22/2022]
Abstract
The circadian timing system orchestrates daily variations in physiology and behavior through coordination of multioscillatory cell networks that are highly plastic in responding to environmental changes. Over the last decade, it has become clear that this plasticity involves structural changes and that the changes may be observed not only in central brain regions where the master clock cells reside but also in clock-controlled structures. This review considers experimental data in invertebrate and vertebrate model systems, mainly flies and mammals, illustrating various forms of structural circadian plasticity from cellular to circuit-based levels. It highlights the importance of these plastic events in the functional adaptation of the clock to the changing environment.
Collapse
Affiliation(s)
- Olivier Bosler
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France.
| | - Clémence Girardet
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France.
| | - Jean-Louis Franc
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France
| | - Denis Becquet
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France
| | - Anne-Marie François-Bellan
- Aix-Marseille Université, CNRS, CRN2M, UMR 7286, Faculté de médecine, secteur nord, Boulevard Pierre Dramard, CS 80011, F-13344 Marseille cedex 15, France
| |
Collapse
|
42
|
Abstract
Arcuate AgRP neurons are critical for food intake. Two pathways leading to AgRP neuron activation and food intake include regulation by peripheral hormones leptin and ghrelin, and neuronal regulation via glutamatergic inputs. In a recent article in Cell Reports, Yang et al. demonstrate 'a third way,' regulation by resident astrocytes.
Collapse
Affiliation(s)
- Nathan C Bingham
- Division of Pediatric Endocrinology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Roger D Cone
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
43
|
Sáez-Orellana F, Godoy PA, Silva-Grecchi T, Barra KM, Fuentealba J. Modulation of the neuronal network activity by P2X receptors and their involvement in neurological disorders. Pharmacol Res 2015; 101:109-15. [PMID: 26122853 DOI: 10.1016/j.phrs.2015.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 11/16/2022]
Abstract
ATP is a key energetic molecule, fundamental to cell function, which also has an important role in the extracellular milieu as a signaling molecule, acting as a chemoattractant for immune cells and as a neuro- and gliotransmitter. The ionotropic P2X receptors are members of an ATP-gated ion channels family. These ionotropic receptors are widely expressed through the body, with 7 subunits described in mammals, which are arranged in a trimeric configuration with a central pore permeable mainly to Ca(2+) and Na(+). All 7 subunits are expressed in different brain areas, being present in neurons and glia. ATP, through these ionotropic receptors, can act as a neuromodulator, facilitating the Ca(2+)-dependent release of neurotransmitters, inducing the cross-inhibition between P2XR and GABA receptors, and exercising by this way a modulation of synaptic plasticity. Growing evidence shows that P2XR play an important role in neuronal disorders and neurodegenerative diseases, like Parkinson's and Alzheimer's disease; this role involves changes on P2XR expression levels, activation of key pathways like GSK3β, APP processing, oxidative stress and inflammatory response. This review is focused on the neuromodulatory function of P2XR on pathophysiological conditions of the brain; the recent evidence could open a window to a new therapeutic target.
Collapse
Affiliation(s)
- F Sáez-Orellana
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile
| | - P A Godoy
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile
| | - T Silva-Grecchi
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile
| | - K M Barra
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile
| | - J Fuentealba
- Screening of Neuroactive Compounds Unit, Department of Physiology, Faculty of Biological Sciences, Chile; Center for Advanced Research on Biomedicine (CIAB-UdeC), University of Concepción, Chile.
| |
Collapse
|
44
|
de Kloet AD, Liu M, Rodríguez V, Krause EG, Sumners C. Role of neurons and glia in the CNS actions of the renin-angiotensin system in cardiovascular control. Am J Physiol Regul Integr Comp Physiol 2015; 309:R444-58. [PMID: 26084692 DOI: 10.1152/ajpregu.00078.2015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/15/2015] [Indexed: 02/07/2023]
Abstract
Despite tremendous research efforts, hypertension remains an epidemic health concern, leading often to the development of cardiovascular disease. It is well established that in many instances, the brain plays an important role in the onset and progression of hypertension via activation of the sympathetic nervous system. Further, the activity of the renin-angiotensin system (RAS) and of glial cell-mediated proinflammatory processes have independently been linked to this neural control and are, as a consequence, both attractive targets for the development of antihypertensive therapeutics. Although it is clear that the predominant effector peptide of the RAS, ANG II, activates its type-1 receptor on neurons to mediate some of its hypertensive actions, additional nuances of this brain RAS control of blood pressure are constantly being uncovered. One of these complexities is that the RAS is now thought to impact cardiovascular control, in part, via facilitating a glial cell-dependent proinflammatory milieu within cardiovascular control centers. Another complexity is that the newly characterized antihypertensive limbs of the RAS are now recognized to, in many cases, antagonize the prohypertensive ANG II type 1 receptor (AT1R)-mediated effects. That being said, the mechanism by which the RAS, glia, and neurons interact to regulate blood pressure is an active area of ongoing research. Here, we review the current understanding of these interactions and present a hypothetical model of how these exchanges may ultimately regulate cardiovascular function.
Collapse
Affiliation(s)
- Annette D de Kloet
- Department of Physiology and Functional Genomics, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida; and
| | - Meng Liu
- Department of Physiology and Functional Genomics, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida; and
| | - Vermalí Rodríguez
- Department of Physiology and Functional Genomics, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida; and
| | - Eric G Krause
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida
| | - Colin Sumners
- Department of Physiology and Functional Genomics, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida; and
| |
Collapse
|
45
|
Stern JE. Neuroendocrine-autonomic integration in the paraventricular nucleus: novel roles for dendritically released neuropeptides. J Neuroendocrinol 2015; 27:487-97. [PMID: 25546497 PMCID: PMC4447596 DOI: 10.1111/jne.12252] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/09/2014] [Accepted: 12/18/2014] [Indexed: 12/21/2022]
Abstract
Communication between pairs of neurones in the central nervous system typically involves classical 'hard-wired' synaptic transmission, characterised by high temporal and spatial precision. Over the last two decades, however, knowledge regarding the repertoire of communication modalities used in the brain has notably expanded to include less conventional forms, characterised by a diffuse and less temporally precise transfer of information. These forms are best suited to mediate communication among entire neuronal populations, now recognised to be a fundamental process in the brain for the generation of complex behaviours. In response to an osmotic stressor, the hypothalamic paraventricular nucleus (PVN) generates a multimodal homeostatic response that involves orchestrated neuroendocrine (i.e. systemic release of vasopressin) and autonomic (i.e. sympathetic outflow to the kidneys) components. The precise mechanisms that underlie interpopulation cross-talk between these two distinct neuronal populations, however, remain largely unknown. The present review summarises and discusses a series of recent studies that have identified the dendritic release of neuropeptides as a novel interpopulation signalling modality in the PVN. A current working model is described in which it is proposed that the activity-dependent dendritic release of vasopressin from neurosecretory neurones in the PVN acts in a diffusible manner to increase the activity of distant presympathetic neurones, resulting in an integrated sympathoexcitatory population response, particularly within the context of a hyperosmotic challenge. The cellular mechanism underlying this novel form of intercellular communication, as well as its physiological and pathophysiological implications, is discussed.
Collapse
Affiliation(s)
- J E Stern
- Department of Physiology, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
46
|
Abstract
Sodium (Na) homeostasis is crucial for life, and the Na(+) level ([Na(+)]) of body fluids is strictly maintained at a range of 135-145 mM. However, the existence of a [Na(+)] sensor in the brain has long been controversial until Nax was identified as the molecular entity of the sensor. This review provides an overview of the [Na(+)]-sensing mechanism in the brain for the regulation of salt intake by summarizing a series of our studies on Nax. Nax is a Na channel expressed in the circumventricular organs (CVOs) in the brain. Among the CVOs, the subfornical organ (SFO) is the principal site for the control of salt intake behavior, where Nax populates the cellular processes of astrocytes and ependymal cells enveloping neurons. A local expression of endothelin-3 in the SFO modulates the [Na(+)] sensitivity for Nax activation, and thereby Nax is likely to be activated in the physiological [Na(+)] range. Nax stably interacts with Na(+)/K(+)-ATPase whereby Na(+) influx via Nax is coupled with activation of Na(+)/K(+)-ATPase associated with the consumption of ATP. The consequent activation of anaerobic glucose metabolism of Nax-positive glial cells upregulates the cellular release of lactate, and this lactate functions as a gliotransmitter to activate GABAergic neurons in the SFO. The GABAergic neurons presumably regulate hypothetic neurons involved in the control of salt intake behavior. Recently, a patient with essential hypernatremia caused by autoimmunity to Nax was found. In this case, the hypernatremia was considered to be induced by the complement-mediated cell death in the CVOs, where Nax specifically populates.
Collapse
Affiliation(s)
- Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology, 5-1 Higashiyama, Myodaiji-cho, Okazaki, 444-8787, Japan,
| | | |
Collapse
|
47
|
Argente-Arizón P, Freire-Regatillo A, Argente J, Chowen JA. Role of non-neuronal cells in body weight and appetite control. Front Endocrinol (Lausanne) 2015; 6:42. [PMID: 25859240 PMCID: PMC4374626 DOI: 10.3389/fendo.2015.00042] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022] Open
Abstract
The brain is composed of neurons and non-neuronal cells, with the latter encompassing glial, ependymal and endothelial cells, as well as pericytes and progenitor cells. Studies aimed at understanding how the brain operates have traditionally focused on neurons, but the importance of non-neuronal cells has become increasingly evident. Once relegated to supporting roles, it is now indubitable that these diverse cell types are fundamental for brain development and function, including that of metabolic circuits, and they may play a significant role in obesity onset and complications. They participate in processes of neurogenesis, synaptogenesis, and synaptic plasticity of metabolic circuits both during development and in adulthood. Some glial cells, such as tanycytes and astrocytes, transport circulating nutrients and metabolic factors that are fundamental for neuronal viability and activity into and within the hypothalamus. All of these cell types express receptors for a variety of metabolic factors and hormones, suggesting that they participate in metabolic function. They are the first line of defense against any assault to neurons. Indeed, microglia and astrocytes participate in the hypothalamic inflammatory response to high fat diet (HFD)-induced obesity, with this process contributing to inflammatory-related insulin and leptin resistance. Moreover, HFD-induced obesity and hyperleptinemia modify hypothalamic astroglial morphology, which is associated with changes in the synaptic inputs to neuronal metabolic circuits. Astrocytic contact with the microvasculature is increased by HFD intake and this could modify nutrient/hormonal uptake into the brain. In addition, progenitor cells in the hypothalamus are now known to have the capacity to renew metabolic circuits, and this can be affected by HFD intake and obesity. Here, we discuss our current understanding of how non-neuronal cells participate in physiological and physiopathological metabolic control.
Collapse
Affiliation(s)
- Pilar Argente-Arizón
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Fisiopatología de la Obesidad y Nutrición (CIBERobn), Centros de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Fisiopatología de la Obesidad y Nutrición (CIBERobn), Centros de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Fisiopatología de la Obesidad y Nutrición (CIBERobn), Centros de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Fisiopatología de la Obesidad y Nutrición (CIBERobn), Centros de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Julie A. Chowen, Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Avda. Menéndez Pelayo, 65, Madrid E-28009, Spain e-mail: ;
| |
Collapse
|
48
|
Vatta MS, Bianciotti LG, Guil MJ, Hope SI. Regulation of the Norepinephrine Transporter by Endothelins. HORMONES AND TRANSPORT SYSTEMS 2015; 98:371-405. [DOI: 10.1016/bs.vh.2014.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
49
|
Brown A, Jordan S. Active management of the third stage of labor may reduce breastfeeding duration due to pain and physical complications. Breastfeed Med 2014; 9:494-502. [PMID: 25347567 DOI: 10.1089/bfm.2014.0048] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Evidence is growing that active management of the third stage of labor using prophylactic uterotonics may be associated with lower breastfeeding rates. The reasons underlying this relationship are incompletely understood. The aim of this article is to examine the experiences of mothers who stopped breastfeeding in relation to administration of parenteral uterotonics for postpartum hemorrhage prophylaxis. SUBJECTS AND METHODS Two hundred eighty-eight mothers with an infant 0-6 months of age who had a vaginal birth completed a self-report questionnaire examining injections of uterotonics during the third stage of labor, breastfeeding at birth, breastfeeding duration, and, where applicable, reasons for breastfeeding cessation, whether physical, social, or psychological. RESULTS No significant association was found between infant feeding mode at birth (breast/formula) and injection of uterotonics. However, mothers who had received uterotonics were significantly less likely to be breastfeeding at all at 2 and 6 weeks. Among mothers who had stopped breastfeeding, those who had received parenteral prophylactic uterotonics were significantly more likely to report stopping breastfeeding for physical reasons such as pain or difficulty. CONCLUSIONS These findings suggest that injection of prophylactic uteronics may reduce breastfeeding duration, but not initiation. This may be attributable to the effects of oxytocin or ergometrine on the physiology of lactation, leading to difficulties with infant latch and milk supply. If breastfeeding rates are to be optimized, this hypothesis needs to be explored in randomized controlled trials of third-stage management. Meanwhile, mothers who receive parenteral uterotonics may need additional support to establish breastfeeding.
Collapse
Affiliation(s)
- Amy Brown
- College of Human and Health Sciences, Swansea University , Swansea, United Kingdom
| | | |
Collapse
|
50
|
Buckman LB, Ellacott KLJ. The contribution of hypothalamic macroglia to the regulation of energy homeostasis. Front Syst Neurosci 2014; 8:212. [PMID: 25374514 PMCID: PMC4206078 DOI: 10.3389/fnsys.2014.00212] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 10/07/2014] [Indexed: 11/13/2022] Open
Abstract
The hypothalamus is critical for the regulation of energy homeostasis. Genetic and pharmacologic studies have identified a number of key hypothalamic neuronal circuits that integrate signals controlling food intake and energy expenditure. Recently, studies have begun to emerge demonstrating a role for non-neuronal cell types in the regulation of energy homeostasis. In particular the potential importance of different glial cell types is increasingly being recognized. A number of studies have described changes in the activity of hypothalamic macroglia (principally astrocytes and tanycytes) in response to states of positive and negative energy balance, such as obesity and fasting. This article will review these studies and discuss how these findings are changing our understanding of the cellular mechanisms by which energy homeostasis is regulated.
Collapse
Affiliation(s)
- Laura B Buckman
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center Nashville, TN, USA ; Division of Infectious Disease, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - Kate L J Ellacott
- Biomedical Neuroscience Research Group, University of Exeter Medical School, Hatherly Laboratories Exeter, UK
| |
Collapse
|