1
|
Ren S, Wang S, Lv S, Gao J, Mao Y, Liu Y, Xie Q, Zhang T, Zhao L, Shi J. The nociceptive inputs of the paraventricular hypothalamic nucleus in formalin stimulated mice. Neurosci Lett 2024; 841:137948. [PMID: 39179131 DOI: 10.1016/j.neulet.2024.137948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/03/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
The paraventricular hypothalamic nucleus (PVH) is an important neuroendocrine center involved in pain regulation, but the nociceptive afferent routes for the nucleus are still unclear. We examined the profile of PVH receiving injurious information by a combination of retrograde tracing with Fluoro-Gold (FG) and FOS expression induced by formalin stimuli. The result showed that formalin injection induced significantly increased expression of FOS in the PVH, among which oxytocin containing neurons are one neuronal phenotype. Immunofluorescent staining of FG and FOS revealed that double labeled neurons were strikingly distributed in the area 2 of the cingulate cortex (Cg2), the lateral septal nucleus (LS), the periaqueductal gray (PAG), the posterior hypothalamic area (PH), and the lateral parabrachial nucleus (LPB). In the five regions, LPB had the biggest number and the highest ratio of FOS expression in FG labeled neurons, with main subnuclei distribution in the external, superior, dorsal, and central parts. Further immunofluorescent triple staining disclosed that about one third of FG and FOS double labeled neurons in the LPB were immunoreactive for calcitonin gene related peptide (CGRP). In conclusion, the present study demonstrates the nociceptive input profile of the PVH area under inflammatory pain and suggests that neurons in the LPB may play essential roles in transmitting noxious information to the PVH.
Collapse
Affiliation(s)
- Shuting Ren
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Medical School of Yan'an University, Yan'an 716000, China
| | - Shumin Wang
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Medical School of Yan'an University, Yan'an 716000, China
| | - Siting Lv
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Medical School of Yan'an University, Yan'an 716000, China
| | - Jiaying Gao
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Yajie Mao
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Yuankun Liu
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Medical School of Yan'an University, Yan'an 716000, China
| | - Qiongyao Xie
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Ting Zhang
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Lin Zhao
- Medical School of Yan'an University, Yan'an 716000, China.
| | - Juan Shi
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
2
|
Verma H, Gangwar P, Yadav A, Yadav B, Rao R, Kaur S, Kumar P, Dhiman M, Taglialatela G, Mantha AK. Understanding the neuronal synapse and challenges associated with the mitochondrial dysfunction in mild cognitive impairment and Alzheimer's disease. Mitochondrion 2023; 73:19-29. [PMID: 37708950 DOI: 10.1016/j.mito.2023.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/26/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023]
Abstract
Synaptic mitochondria are crucial for maintaining synaptic activity due to their high energy requirements, substantial calcium (Ca2+) fluctuation, and neurotransmitter release at the synapse. To provide a continuous energy supply, neurons use special mechanisms to transport and distribute healthy mitochondria to the synapse while eliminating the damaged mitochondria from the synapse. Along the neuron, mitochondrial membrane potential (ψ) gradient exists and is highest in the somal region. Lower ψ in the synaptic region renders mitochondria more vulnerable to oxidative stress-mediated damage. Secondly, mitochondria become susceptible to the release of cytochrome c, and mitochondrial DNA (mtDNA) is not shielded from the reactive oxygen species (ROS) by the histone proteins (unlike nuclear DNA), leading to activation of caspases and pronounced oxidative DNA base damage, which ultimately causes synaptic loss. Both synaptic mitochondrial dysfunction and synaptic failure are crucial factors responsible for Alzheimer's disease (AD). Furthermore, amyloid beta (Aβ) and hyper-phosphorylated Tau, the two leading players of AD, exaggerate the disease-like pathological conditions by reducing the mitochondrial trafficking, blocking the bi-directional transport at the synapse, enhancing the mitochondrial fission via activating the mitochondrial fission proteins, enhancing the swelling of mitochondria by increasing the influx of water through mitochondrial permeability transition pore (mPTP) opening, as well as reduced ATP production by blocking the activity of complex I and complex IV. Mild cognitive impairment (MCI) is also associated with decline in cognitive ability caused by synaptic degradation. This review summarizes the challenges associated with the synaptic mitochondrial dysfunction linked to AD and MCI and the role of phytochemicals in restoring the synaptic activity and rendering neuroprotection in AD.
Collapse
Affiliation(s)
- Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Prabhakar Gangwar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Anuradha Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Bharti Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Rashmi Rao
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Giulio Taglialatela
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Anil Kumar Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India.
| |
Collapse
|
3
|
Greenwood MP, Greenwood M, Bárez-López S, Hawkins JW, Short K, Tatovic D, Murphy D. Osmoadaptive GLP-1R signalling in hypothalamic neurones inhibits antidiuretic hormone synthesis and release. Mol Metab 2023; 70:101692. [PMID: 36773648 PMCID: PMC9969259 DOI: 10.1016/j.molmet.2023.101692] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
OBJECTIVES The excessive release of the antidiuretic hormone vasopressin is implicated in many diseases including cardiovascular disease, diabetes, obesity, and metabolic syndrome. Once thought to be elevated as a consequence of diseases, data now supports a more causative role. We have previously identified CREB3L1 as a transcription factor that co-ordinates vasopressin synthesis and release in the hypothalamus. The objective here was to identify mechanisms orchestrated by CREB3L1 that co-ordinate vasopressin release. METHODS We mined Creb3l1 knockdown SON RNA-seq data to identify downstream target genes. We proceeded to investigate the expression of these genes and associated pathways in the supraoptic nucleus of the hypothalamus in response to physiological and pharmacological stimulation. We used viruses to selectively knockdown gene expression in the supraoptic nucleus and assessed physiological and metabolic parameters. We adopted a phosphoproteomics strategy to investigate mechanisms that facilitate hormone release by the pituitary gland. RESULTS We discovered glucagon like peptide 1 receptor (Glp1r) as a downstream target gene and found increased expression in stimulated vasopressin neurones. Selective knockdown of supraoptic nucleus Glp1rs resulted in decreased food intake and body weight. Treatment with GLP-1R agonist liraglutide decreased vasopressin synthesis and release. Quantitative phosphoproteomics of the pituitary neurointermediate lobe revealed that liraglutide initiates hyperphosphorylation of presynapse active zone proteins that control vasopressin exocytosis. CONCLUSION In summary, we show that GLP-1R signalling inhibits the vasopressin system. Our data advises that hydration status may influence the pharmacodynamics of GLP-1R agonists so should be considered in current therapeutic strategies.
Collapse
Affiliation(s)
- Michael P Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom.
| | - Mingkwan Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - Soledad Bárez-López
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - Joe W Hawkins
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - Katherine Short
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - Danijela Tatovic
- Diabetes and Endocrinology Department, North Bristol NHS Trust, Bristol, United Kingdom
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| |
Collapse
|
4
|
Martucci LL, Launay JM, Kawakami N, Sicard C, Desvignes N, Dakouane-Giudicelli M, Spix B, Têtu M, Gilmaire FO, Paulcan S, Callebert J, Vaillend C, Bracher F, Grimm C, Fossier P, de la Porte S, Sakamoto H, Morris J, Galione A, Granon S, Cancela JM. Endolysosomal TPCs regulate social behavior by controlling oxytocin secretion. Proc Natl Acad Sci U S A 2023; 120:e2213682120. [PMID: 36745816 PMCID: PMC9963339 DOI: 10.1073/pnas.2213682120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/14/2022] [Indexed: 02/08/2023] Open
Abstract
Oxytocin (OT) is a prominent regulator of many aspects of mammalian social behavior and stored in large dense-cored vesicles (LDCVs) in hypothalamic neurons. It is released in response to activity-dependent Ca2+ influx, but is also dependent on Ca2+ release from intracellular stores, which primes LDCVs for exocytosis. Despite its importance, critical aspects of the Ca2+-dependent mechanisms of its secretion remain to be identified. Here we show that lysosomes surround dendritic LDCVs, and that the direct activation of endolysosomal two-pore channels (TPCs) provides the critical Ca2+ signals to prime OT release by increasing the releasable LDCV pool without directly stimulating exocytosis. We observed a dramatic reduction in plasma OT levels in TPC knockout mice, and impaired secretion of OT from the hypothalamus demonstrating the importance of priming of neuropeptide vesicles for activity-dependent release. Furthermore, we show that activation of type 1 metabotropic glutamate receptors sustains somatodendritic OT release by recruiting TPCs. The priming effect could be mimicked by a direct application of nicotinic acid adenine dinucleotide phosphate, the endogenous messenger regulating TPCs, or a selective TPC2 agonist, TPC2-A1-N, or blocked by the antagonist Ned-19. Mice lacking TPCs exhibit impaired maternal and social behavior, which is restored by direct OT administration. This study demonstrates an unexpected role for lysosomes and TPCs in controlling neuropeptide secretion, and in regulating social behavior.
Collapse
Affiliation(s)
- Lora L. Martucci
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
- Université Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines, Inserm, Evolution of Neuromuscular Diseases: Innovative Concepts and Practices, Versailles78000, France
- Department of Pharmacology, University of Oxford, OxfordOX1 3QT, UK
| | | | - Natsuko Kawakami
- Ushimado Marine Institute, Graduate School of Natural Science and Technology, Okayama University, Ushimado, Setouchi, Okayama701-4303, Japan
| | - Cécile Sicard
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Nathalie Desvignes
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Mbarka Dakouane-Giudicelli
- Université Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines, Inserm, Evolution of Neuromuscular Diseases: Innovative Concepts and Practices, Versailles78000, France
| | - Barbara Spix
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine Ludwig-Maximilians-University, Munich80336, Germany
| | - Maude Têtu
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Franck-Olivier Gilmaire
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Sloane Paulcan
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Jacques Callebert
- Laboratoire de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, Paris75010, France
- Inserm UMR-S 1144 Universités Paris Descartes-Paris Diderot, Optimisation Thérapeutique en Neuropsychopharmacologie - Faculté des Sciences Pharmaceutiques et Biologiques, Paris Descartes,ParisParis 75006, France
| | - Cyrille Vaillend
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Franz Bracher
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians University, Munich81377, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine Ludwig-Maximilians-University, Munich80336, Germany
| | - Philippe Fossier
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Sabine de la Porte
- Université Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines, Inserm, Evolution of Neuromuscular Diseases: Innovative Concepts and Practices, Versailles78000, France
| | - Hirotaka Sakamoto
- Ushimado Marine Institute, Graduate School of Natural Science and Technology, Okayama University, Ushimado, Setouchi, Okayama701-4303, Japan
| | - John Morris
- Department of Physiology, Anatomy & Genetics, University of Oxford, OxfordOX1 3QX, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, OxfordOX1 3QT, UK
| | - Sylvie Granon
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - José-Manuel Cancela
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| |
Collapse
|
5
|
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
|
6
|
Angelousi A, Alexandraki KI, Mytareli C, Grossman AB, Kaltsas G. New developments and concepts in the diagnosis and management of diabetes insipidus (AVP-deficiency and resistance). J Neuroendocrinol 2023; 35:e13233. [PMID: 36683321 DOI: 10.1111/jne.13233] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/10/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023]
Abstract
Diabetes insipidus (DI) is a disorder characterised by the excretion of large amounts of hypotonic urine, with a prevalence of 1 per 25,000 population. Central DI (CDI), better now referred to as arginine vasopressin (AVP)-deficiency, is the most common form of DI resulting from deficiency of the hormone AVP from the pituitary. The less common nephrogenic DI (NDI) or AVP-resistance develops secondary to AVP resistance in the kidneys. The majority of causes of DI are acquired, with CDI developing when more than 80% of AVP-secreting neurons are damaged. Inherited/familial CDI causes account for approximately 1% of cases. Although the pathogenesis of NDI is unclear, more than 280 disease-causing mutations affecting the AVP2 protein or AVP V2 receptor, as well as in aquaporin 2 (AQP2), have been described. Although the cAMP/protein kinase A pathway remains the major regulatory pathway of AVP/AQP2 action, in vitro data have also revealed additional cAMP independent pathways of NDI pathogenesis. Diagnosing partial forms of DI, and distinguishing them from primary polydipsia, can be challenging, previously necessitating the use of the water deprivation test. However, measurements of circulating copeptin levels, especially after stimulation, are increasingly replacing the classical tests in clinical practice because of their ease of use and high sensitivity and specificity. The treatment of CDI relies on desmopressin administration, whereas NDI requires the management of any underlying diseases, removal of offending drugs and, in some cases, administration of diuretics. A better understanding of the pathophysiology of DI has led to novel evolving therapeutic agents that are under clinical trial.
Collapse
Affiliation(s)
- Anna Angelousi
- First Department of Internal Medicine, Unit of Endocrinology, Laikon Hospital, Athens, Greece
| | | | - Chrysoula Mytareli
- First Department of Internal Medicine, Unit of Endocrinology, Laikon Hospital, Athens, Greece
| | - Ashley B Grossman
- Green Templeton College, University of Oxford, Oxford, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
- NET Unit, Royal Free Hospital, London, UK
| | - Gregory Kaltsas
- First Department of Propaedeutic Internal Medicine, Laikon Hospital, National & Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
7
|
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: 6.0] [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
|
8
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
9
|
Duittoz A, Cayla X, Fleurot R, Lehnert J, Khadra A. Gonadotrophin-releasing hormone and kisspeptin: It takes two to tango. J Neuroendocrinol 2021; 33:e13037. [PMID: 34533248 DOI: 10.1111/jne.13037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/06/2023]
Abstract
Kisspeptin (Kp), a family of peptides comprising products of the Kiss1 gene, was discovered 20 years ago; it is recognised as the major factor controlling the activity of the gonadotrophin-releasing hormone (GnRH) neurones and thus the activation of the reproductive axis in mammals. It has been widely documented that the effects of Kp on reproduction through its action on GnRH neurones are mediated by the GPR54 receptor. Kp controls the activation of the reproductive axis at puberty, maintains reproductive axis activity in adults and is involved in triggering ovulation in some species. Although there is ample evidence coming from both conditional knockout models and conditional-induced Kp neurone death implicating the Kp/GPR54 pathway in the control of reproduction, the mechanism(s) underlying this process may be more complex than a sole direct control of GnRH neuronal activity by Kp. In this review, we provide an overview of the recent advances made in elucidating the interplay between Kp- and GnRH- neuronal networks with respect to regulating the reproductive axis. We highlight the existence of a possible mutual regulation between GnRH and Kp neurones, as well as the implication of Kp-dependent volume transmission in this process. We also discuss the capacity of heterodimerisation between GPR54 and GnRH receptor (GnRH-R) and its consequences on signalling. Finally, we illustrate the role of mathematical modelling that accounts for the synergy between GnRH-R and GPR54 in explaining the role of these two receptors when defining GnRH neuronal activity and GnRH pulsatile release.
Collapse
Affiliation(s)
- Anne Duittoz
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Xavier Cayla
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Renaud Fleurot
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Jonas Lehnert
- Department of Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Anmar Khadra
- Department of Quantitative Life Sciences, McGill University, Montreal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| |
Collapse
|
10
|
Sheng W, Harden SW, Tan Y, Krause EG, Frazier CJ. Dendritic osmosensors modulate activity-induced calcium influx in oxytocinergic magnocellular neurons of the mouse PVN. eLife 2021; 10:e63486. [PMID: 34250900 PMCID: PMC8457833 DOI: 10.7554/elife.63486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 07/11/2021] [Indexed: 11/13/2022] Open
Abstract
Hypothalamic oxytocinergic magnocellular neurons have a fascinating ability to release peptide from both their axon terminals and from their dendrites. Existing data indicates that the relationship between somatic activity and dendritic release is not constant, but the mechanisms through which this relationship can be modulated are not completely understood. Here, we use a combination of electrical and optical recording techniques to quantify activity-induced calcium influx in proximal vs. distal dendrites of oxytocinergic magnocellular neurons located in the paraventricular nucleus of the hypothalamus (OT-MCNs). Results reveal that the dendrites of OT-MCNs are weak conductors of somatic voltage changes; however, activity-induced dendritic calcium influx can be robustly regulated by both osmosensitive and non-osmosensitive ion channels located along the dendritic membrane. Overall, this study reveals that dendritic conductivity is a dynamic and endogenously regulated feature of OT-MCNs that is likely to have substantial functional impact on central oxytocin release.
Collapse
Affiliation(s)
- Wanhui Sheng
- Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesvilleUnited States
| | - Scott W Harden
- Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesvilleUnited States
| | - Yalun Tan
- Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesvilleUnited States
- Department of Anesthesiology, School of Medicine, Stanford UniversityStanfordUnited States
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesvilleUnited States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of FloridaGainesvilleUnited States
- Evelyn F. and William L. McKnight Brain Institute, University of FloridaGainesvilleUnited States
| | - Charles J Frazier
- Department of Pharmacodynamics, College of Pharmacy, University of FloridaGainesvilleUnited States
- Center for Integrative Cardiovascular and Metabolic Diseases, University of FloridaGainesvilleUnited States
| |
Collapse
|
11
|
De-Miguel FF, Leon-Pinzon C, Torres-Platas SG, Del-Pozo V, Hernández-Mendoza GA, Aguirre-Olivas D, Méndez B, Moore S, Sánchez-Sugía C, García-Aguilera MA, Martínez-Valencia A, Ramírez-Santiago G, Rubí JM. Extrasynaptic Communication. Front Mol Neurosci 2021; 14:638858. [PMID: 33994942 PMCID: PMC8119753 DOI: 10.3389/fnmol.2021.638858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Streams of action potentials or long depolarizations evoke a massive exocytosis of transmitters and peptides from the surface of dendrites, axons and cell bodies of different neuron types. Such mode of exocytosis is known as extrasynaptic for occurring without utilization of synaptic structures. Most transmitters and all peptides can be released extrasynaptically. Neurons may discharge their contents with relative independence from the axon, soma and dendrites. Extrasynaptic exocytosis takes fractions of a second in varicosities or minutes in the soma or dendrites, but its effects last from seconds to hours. Unlike synaptic exocytosis, which is well localized, extrasynaptic exocytosis is diffuse and affects neuronal circuits, glia and blood vessels. Molecules that are liberated may reach extrasynaptic receptors microns away. The coupling between excitation and exocytosis follows a multistep mechanism, different from that at synapses, but similar to that for the release of hormones. The steps from excitation to exocytosis have been studied step by step for the vital transmitter serotonin in leech Retzius neurons. The events leading to serotonin exocytosis occur similarly for the release of other transmitters and peptides in central and peripheral neurons. Extrasynaptic exocytosis occurs commonly onto glial cells, which react by releasing the same or other transmitters. In the last section, we discuss how illumination of the retina evokes extrasynaptic release of dopamine and ATP. Dopamine contributes to light-adaptation; ATP activates glia, which mediates an increase in blood flow and oxygenation. A proper understanding of the workings of the nervous system requires the understanding of extrasynaptic communication.
Collapse
Affiliation(s)
- Francisco F De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, México City, Mexico
| | - Carolina Leon-Pinzon
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Susana G Torres-Platas
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Vanessa Del-Pozo
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | | | - Dilia Aguirre-Olivas
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Bruno Méndez
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Sharlen Moore
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | - Celeste Sánchez-Sugía
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, México City, Mexico
| | | | | | | | - J Miguel Rubí
- Facultat de Fisica, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
MacGregor DJ, Leng G. Network and Population Function in Neuroendocrine Systems. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11371-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
13
|
García-Ávila M, Torres X, Cercós MG, Trueta C. Specific Localization of an Auto-inhibition Mechanism at Presynaptic Terminals of Identified Serotonergic Neurons. Neuroscience 2020; 458:120-132. [PMID: 33359652 DOI: 10.1016/j.neuroscience.2020.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 11/18/2022]
Abstract
Auto-regulation mechanisms in serotonergic neurons regulate their electrical activity and secretion. Since these neurons release serotonin from different structural compartments - including presynaptic terminals, soma, axons and dendrites - through different mechanisms, autoregulation mechanisms are also likely to be different at each compartment. Here we show that a chloride-mediated auto-inhibitory mechanism is exclusively localized at presynaptic terminals, but not at extrasynaptic release sites, in serotonergic Retzius neurons of the leech. An auto-inhibition response was observed immediately after intracellular stimulation with an electrode placed in the soma, in neurons that were isolated and cultured retaining an axonal stump, where presynaptic terminals are formed near the soma, but not in somata isolated without axon, where no synaptic terminals are formed, nor in neurons in the nerve ganglion, where terminals are electrotonically distant from the soma. Furthermore, no auto-inhibition response was detected in either condition during the longer time course of somatic secretion. This shows that the auto-inhibition effects are unique to nerve terminals. We further determined that serotonin released from peri-synaptic dense-core vesicles contributes to auto-inhibition in the terminals, since blockade of L-type calcium channels, which are required to stimulate extrasynaptic but not synaptic release, decreased the amplitude of the auto-inhibition response. Our results show that the auto-regulation mechanism at presynaptic terminals is unique and different from that described in the soma of these neurons, further highlighting the differences in the mechanisms regulating serotonin release from different neuronal compartments, which expand the possibilities of a single neuron to perform multiple functions in the nervous system.
Collapse
Affiliation(s)
- Miriam García-Ávila
- Departamento de Neurofisiología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, San Lorenzo Huipulco, Tlalpan 14370, Ciudad de México, Mexico.
| | - Ximena Torres
- Departamento de Neurofisiología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, San Lorenzo Huipulco, Tlalpan 14370, Ciudad de México, Mexico.
| | - Montserrat G Cercós
- Departamento de Neurofisiología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, San Lorenzo Huipulco, Tlalpan 14370, Ciudad de México, Mexico.
| | - Citlali Trueta
- Departamento de Neurofisiología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, San Lorenzo Huipulco, Tlalpan 14370, Ciudad de México, Mexico.
| |
Collapse
|
14
|
Kuo C, Foon D, Waters K, Cheung C, Margol AS. Central diabetes insipidus: A rare unreported side effect of temozolomide in pediatrics. Pediatr Blood Cancer 2020; 67:e28516. [PMID: 32573959 DOI: 10.1002/pbc.28516] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Christopher Kuo
- Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California
| | - Dione Foon
- Cancer and Blood Disease Institute and Division of Hematology Oncology, Children's Hospital Los Angeles, Los Angeles, California
| | - Kaaren Waters
- Cancer and Blood Disease Institute and Division of Hematology Oncology, Children's Hospital Los Angeles, Los Angeles, California
| | - Clement Cheung
- Division of Endocrinology, Diabetes, and Metabolism, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California.,Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ashley S Margol
- Cancer and Blood Disease Institute and Division of Hematology Oncology, Children's Hospital Los Angeles, Los Angeles, California.,Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
15
|
Mahiat C, Capes A, Duprez T, Whenham N, Duck L, Labriola L. Central diabetes insipidus induced by temozolomide: A report of two cases. J Oncol Pharm Pract 2020; 27:1040-1045. [PMID: 32990192 DOI: 10.1177/1078155220961551] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Central diabetes insipidus is a heterogeneous condition characterized by decreased release of antidiuretic hormone by the neurohypophysis resulting in a urine concentration deficit with variable degrees of polyuria. The most common causes include idiopathic diabetes insipidus, tumors or infiltrative diseases, neurosurgery and trauma. Temozolomide is an oral DNA-alkylating agent capable of crossing the blood-brain barrier and used as chemotherapy primarily to treat glioblastoma and other brain cancers. CASES Two men (aged 38 and 54 years) suddenly developed polyuria and polydispsia approximately four weeks after the initiation of temozolomide for a glioblastoma. Plasma and urine parameters demonstrated the presence of a urinary concentration defect. MANAGEMENT The clinical and laboratory abnormalities completely resolved with intranasal desmopressin therapy, allowing the continuation of temozolomide. The disorder did not relapse after cessation of temozolomide and desmopressin and relapsed in one patient after rechallenge with temozolomide. DISCUSSION Our report highlights the importance of a quick recognition of this exceptional complication, in order to initiate promptly treatment with desmopressin and to maintain therapy with temozolomide.
Collapse
Affiliation(s)
- Cédric Mahiat
- Department of Oncology, Clinique Saint-Pierre, Ottignies, Belgium
| | - Antoine Capes
- Department of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Thierry Duprez
- Department of Medical Imaging, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Nicolas Whenham
- Department of Oncology, Clinique Saint-Pierre, Ottignies, Belgium.,Department of Oncology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Lionel Duck
- Department of Oncology, Clinique Saint-Pierre, Ottignies, Belgium
| | - Laura Labriola
- Department of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
16
|
Hagiwara A, Sugiyama N, Ohtsuka T. Impaired experience-dependent maternal care in presynaptic active zone protein CAST-deficient dams. Sci Rep 2020; 10:5238. [PMID: 32251313 PMCID: PMC7090055 DOI: 10.1038/s41598-020-62072-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/09/2020] [Indexed: 11/24/2022] Open
Abstract
Although sociological studies affirm the importance of parental care in the survival of offspring, maltreatment—including child neglect—remains prevalent in many countries. While child neglect is well known to affect child development, the causes of maternal neglect are poorly understood. Here, we found that female mice with a deletion mutation of CAST (a presynaptic release-machinery protein) showed significantly reduced weaning rate when primiparous and a recovered rate when multiparous. Indeed, when nurturing, primiparous and nulliparous CAST knock out (KO) mice exhibited less crouching time than control mice and moved greater distances. Contrary to expectations, plasma oxytocin (OXT) was not significantly reduced in CAST KO mice even though terminals of magnocellular neurons in the posterior pituitary expressed CAST. We further found that compared with control mice, CAST KO mice drank significantly less water when nurturing and had a greater preference for sucrose during pregnancy. We suggest that deficiency in presynaptic release-machinery protein impairs the facilitation of some maternal behaviours, which can be compensated for by experience and learning.
Collapse
Affiliation(s)
- Akari Hagiwara
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Naoko Sugiyama
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan.
| |
Collapse
|
17
|
A Fear Memory Engram and Its Plasticity in the Hypothalamic Oxytocin System. Neuron 2019; 103:133-146.e8. [PMID: 31104950 DOI: 10.1016/j.neuron.2019.04.029] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/08/2018] [Accepted: 04/18/2019] [Indexed: 11/24/2022]
Abstract
Oxytocin (OT) release by axonal terminals onto the central nucleus of the amygdala exerts anxiolysis. To investigate which subpopulation of OT neurons contributes to this effect, we developed a novel method: virus-delivered genetic activity-induced tagging of cell ensembles (vGATE). With the vGATE method, we identified and permanently tagged a small subpopulation of OT cells, which, by optogenetic stimulation, strongly attenuated contextual fear-induced freezing, and pharmacogenetic silencing of tagged OT neurons impaired context-specific fear extinction, demonstrating that the tagged OT neurons are sufficient and necessary, respectively, to control contextual fear. Intriguingly, OT cell terminals of fear-experienced rats displayed enhanced glutamate release in the amygdala. Furthermore, rats exposed to another round of fear conditioning displayed 5-fold more activated magnocellular OT neurons in a novel environment than a familiar one, possibly for a generalized fear response. Thus, our results provide first evidence that hypothalamic OT neurons represent a fear memory engram.
Collapse
|
18
|
Gliotransmission: Beyond Black-and-White. J Neurosci 2019; 38:14-25. [PMID: 29298905 DOI: 10.1523/jneurosci.0017-17.2017] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/01/2017] [Accepted: 08/29/2017] [Indexed: 01/09/2023] Open
Abstract
Astrocytes are highly complex cells with many emerging putative roles in brain function. Of these, gliotransmission (active information transfer from glia to neurons) has probably the widest implications on our understanding of how the brain works: do astrocytes really contribute to information processing within the neural circuitry? "Positive evidence" for this stems from work of multiple laboratories reporting many examples of modulatory chemical signaling from astrocytes to neurons in the timeframe of hundreds of milliseconds to several minutes. This signaling involves, but is not limited to, Ca2+-dependent vesicular transmitter release, and results in a variety of regulatory effects at synapses in many circuits that are abolished by preventing Ca2+ elevations or blocking exocytosis selectively in astrocytes. In striking contradiction, methodologically advanced studies by a few laboratories produced "negative evidence," triggering a heated debate on the actual existence and properties of gliotransmission. In this context, a skeptics' camp arose, eager to dismiss the whole positive evidence based on a number of assumptions behind the negative data, such as the following: (1) deleting a single Ca2+ release pathway (IP3R2) removes all the sources for Ca2+-dependent gliotransmission; (2) stimulating a transgenically expressed Gq-GPCR (MrgA1) mimics the physiological Ca2+ signaling underlying gliotransmitter release; (3) age-dependent downregulation of an endogenous GPCR (mGluR5) questions gliotransmitter release in adulthood; and (4) failure by transcriptome analysis to detect vGluts or canonical synaptic SNAREs in astrocytes proves inexistence/functional irrelevance of vesicular gliotransmitter release. We here discuss how the above assumptions are likely wrong and oversimplistic. In light of the most recent literature, we argue that gliotransmission is a more complex phenomenon than originally thought, possibly consisting of multiple forms and signaling processes, whose correct study and understanding require more sophisticated tools and finer scientific experiments than done until today. Under this perspective, the opposing camps can be reconciled and the field moved forward. Along the path, a more cautious mindset and an attitude to open discussion and mutual respect between opponent laboratories will be good companions.Dual Perspectives Companion Paper: Multiple Lines of Evidence Indicate That Gliotransmission Does Not Occur under Physiological Conditions, by Todd A. Fiacco and Ken D. McCarthy.
Collapse
|
19
|
Expanding Regulation Theory With Oxytocin: A Psychoneurobiological Model for Infant Development. Nurs Res 2019; 67:133-145. [PMID: 29489634 DOI: 10.1097/nnr.0000000000000261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Oxytocin (OT), an affiliation hormone released during supportive social interactions, provides an exemplar of how social environments are reflected in our neurobiology from the beginning of life. A growing body of OT research has uncovered previously unknown functions of OT, including modulation of parenting behaviors, neuroprotection, affiliation, and bonding. Regulation theory provides a strong framework for describing how the maternal care environment affects infant neurodevelopment through a symphony of molecules that form the neurobiological milieu of the developing infant brain. OBJECTIVES The purpose of this article was to expand on regulation theory by discussing how OT-based processes contribute to infant neurobiology and by proposing a new model for maternal-infant nursing practice and research. APPROACH We structure our discussion of the socially based, neurobiological processes of OT through its effects in the nested hierarchies of genetic, epigenetic, molecular, cellular, neural circuit, multiorgan, and behavioral levels. Our discussion is also presented chronologically, as OT works through a positive feedback loop during infant neurodevelopment, beginning prenatally and continuing after birth. IMPLICATIONS Nurses are in a unique position to use innovative discoveries made by the biologic sciences to generate new nursing theories that inform clinical practice and inspire the development of innovative interventions that maximize the infant's exposure to supportive maternal care.
Collapse
|
20
|
Erdozain AM, Peñagarikano O. Oxytocin as Treatment for Social Cognition, Not There Yet. Front Psychiatry 2019; 10:930. [PMID: 31998152 PMCID: PMC6962227 DOI: 10.3389/fpsyt.2019.00930] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/22/2019] [Indexed: 12/15/2022] Open
Abstract
In a short time, oxytocin has progressed from being a regular hormone involved in parturition and breastfeeding to be possibly the neuromodulator that has gathered the most attention. Attributed many positive roles in the modulation of different aspects of social behavior, such as bonding, empathy, cooperation, trust, and generosity, as well as roles as a natural anxiolytic and antidepressant, the expectations on oxytocin becoming a treatment for a number of disorders with associated social deficits have dramatically raised over the last years. However, despite the field has been investigating oxytocin's role in social behavior for over twenty years, there are still many unknowns on oxytocin's mechanisms of action and efficiency and the increasing number of clinical trials administering oxytocin to different clinical groups seem to disagree in its properties and report in most cases conflicting results. This has led to some disappointment among researchers and clinicians as oxytocin might not be the miraculous molecule that works in a "one size fits all" fashion initially considered. Conversely, this down-side of oxytocin might merely reflect the complexity of its neurotransmission system. The current reality is that, although oxytocin seems to have potential therapeutic value, there are key questions that remain unanswered as to decide the optimal target groups and treatment course. Here, we present an overview on critical points regarding the oxytocin system in health and disease that need to be better understood to establish its therapeutic properties and to decide who could benefit the most from its treatment.
Collapse
Affiliation(s)
- Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Spain.,Centro de Investigación Biomédica en Red en Salud Mental CIBERSAM, Leioa, Spain
| | - Olga Peñagarikano
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Spain.,Centro de Investigación Biomédica en Red en Salud Mental CIBERSAM, Leioa, Spain
| |
Collapse
|
21
|
Abstract
The brain hosts a vast and diverse repertoire of neuropeptides, a class of signalling molecules often described as neurotransmitters. Here I argue that this description entails a catalogue of misperceptions, misperceptions that feed into a narrative in which information processing in the brain can be understood only through mapping neuronal connectivity and by studying the transmission of electrically conducted signals through chemical synapses. I argue that neuropeptide signalling in the brain involves primarily autocrine, paracrine and neurohormonal mechanisms that do not depend on synaptic connectivity and that it is not solely dependent on electrical activity but on mechanisms analogous to secretion from classical endocrine cells. As in classical endocrine systems, to understand the role of neuropeptides in the brain, we must understand not only how their release is regulated, but also how their synthesis is regulated and how the sensitivity of their targets is regulated. We must also understand the full diversity of effects of neuropeptides on those targets, including their effects on gene expression.
Collapse
Affiliation(s)
- Gareth Leng
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Correspondence should be addressed to G Leng:
| |
Collapse
|
22
|
Leng G, MacGregor DJ. Models in neuroendocrinology. Math Biosci 2018; 305:29-41. [DOI: 10.1016/j.mbs.2018.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/20/2018] [Accepted: 07/24/2018] [Indexed: 12/18/2022]
|
23
|
Jurek B, Neumann ID. The Oxytocin Receptor: From Intracellular Signaling to Behavior. Physiol Rev 2018; 98:1805-1908. [DOI: 10.1152/physrev.00031.2017] [Citation(s) in RCA: 408] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The many facets of the oxytocin (OXT) system of the brain and periphery elicited nearly 25,000 publications since 1930 (see FIGURE 1 , as listed in PubMed), which revealed central roles for OXT and its receptor (OXTR) in reproduction, and social and emotional behaviors in animal and human studies focusing on mental and physical health and disease. In this review, we discuss the mechanisms of OXT expression and release, expression and binding of the OXTR in brain and periphery, OXTR-coupled signaling cascades, and their involvement in behavioral outcomes to assemble a comprehensive picture of the central and peripheral OXT system. Traditionally known for its role in milk let-down and uterine contraction during labor, OXT also has implications in physiological, and also behavioral, aspects of reproduction, such as sexual and maternal behaviors and pair bonding, but also anxiety, trust, sociability, food intake, or even drug abuse. The many facets of OXT are, on a molecular basis, brought about by a single receptor. The OXTR, a 7-transmembrane G protein-coupled receptor capable of binding to either Gαior Gαqproteins, activates a set of signaling cascades, such as the MAPK, PKC, PLC, or CaMK pathways, which converge on transcription factors like CREB or MEF-2. The cellular response to OXT includes regulation of neurite outgrowth, cellular viability, and increased survival. OXTergic projections in the brain represent anxiety and stress-regulating circuits connecting the paraventricular nucleus of the hypothalamus, amygdala, bed nucleus of the stria terminalis, or the medial prefrontal cortex. Which OXT-induced patterns finally alter the behavior of an animal or a human being is still poorly understood, and studying those OXTR-coupled signaling cascades is one initial step toward a better understanding of the molecular background of those behavioral effects.
Collapse
Affiliation(s)
- Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Inga D. Neumann
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
24
|
Rajamani KT, Wagner S, Grinevich V, Harony-Nicolas H. Oxytocin as a Modulator of Synaptic Plasticity: Implications for Neurodevelopmental Disorders. Front Synaptic Neurosci 2018; 10:17. [PMID: 29970997 PMCID: PMC6018411 DOI: 10.3389/fnsyn.2018.00017] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/25/2018] [Indexed: 01/23/2023] Open
Abstract
The neuropeptide oxytocin (OXT) is a crucial mediator of parturition and milk ejection and a major modulator of various social behaviors, including social recognition, aggression and parenting. In the past decade, there has been significant excitement around the possible use of OXT to treat behavioral deficits in neurodevelopmental disorders, including autism spectrum disorder (ASD). Yet, despite the fast move to clinical trials with OXT, little attention has been paid to the possibility that the OXT system in the brain is perturbed in these disorders and to what extent such perturbations may contribute to social behavior deficits. Large-scale whole-exome sequencing studies in subjects with ASD, along with biochemical and electrophysiological studies in animal models of the disorder, indicate several risk genes that play an essential role in brain synapses, suggesting that deficits in synaptic activity and plasticity underlie the pathophysiology in a considerable portion of these cases. OXT has been repeatedly shown, both in vitro and in vivo, to modify synaptic properties and plasticity and to modulate neural activity in circuits that regulate social behavior. Together, these findings led us to hypothesize that failure of the OXT system during early development, as a direct or indirect consequence of genetic mutations, may impact social behavior by altering synaptic activity and plasticity. In this article, we review the evidence that support our hypothesis.
Collapse
Affiliation(s)
- Keerthi Thirtamara Rajamani
- The Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,The Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Shlomo Wagner
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Valery Grinevich
- Schaller Research Group on Neuropeptides at German Cancer Research Center (DKFZ), Central Institute of Mental Health and Cell Networks Cluster of Excellence, University of Heidelberg, Heidelberg, Germany
| | - Hala Harony-Nicolas
- The Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,The Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| |
Collapse
|
25
|
Nakashima K, Takeuchi S, Iwama S, Kiyota A, Yasuda Y, Iwata N, Enomoto A, Arima H, Sugimura Y. Cullin-associated NEDD8-dissociated protein 1, a novel interactor of rabphilin-3A, deubiquitylates rabphilin-3A and regulates arginine vasopressin secretion in PC12 cells. Endocr J 2018; 65:325-334. [PMID: 29367474 DOI: 10.1507/endocrj.ej17-0399] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The molecular mechanism involved in the exocytosis of arginine vasopressin (AVP) is not fully known. Rabphilin-3A has been suggested as a novel autoantigen in infundibulo-neurohypophysitis (LINH), which leads to central diabetes insipidus through insufficient secretion of AVP. However, the role of rabphilin-3A in the pathogenesis of LINH remains unclear. Thus, the aim of the present study was to identify proteins binding rabphilin-3A in the posterior pituitary. Using glutathione S-transferase (GST)-pulldown assays and proteomic analyses, cullin-associated NEDD8-dissociated protein 1 (CAND1) was identified as a rabphilin-3A-binding protein in the posterior pituitary. Co-immunoprecipitation assays indicated that CAND1 interacted endogenously with rabphilin-3A. In addition, immunohistochemistry experiments showed that CAND1 immunoreactivity was detected mainly in the posterior pituitary, intermediate lobe, and the supraoptic nucleus in the hypothalamus, and less in the anterior lobe, partially co-localizing with rabphilin-3A. Overexpression of CAND1 resulted in deubiquitylation of rabphilin-3A in PC12 cells. Moreover, overexpression of CAND1 in PC12 cells co-transfected with AVP enhanced both basal and KCl-stimulated AVP secretion. The findings indicate that CAND1 inhibits the ubiquitylation of rabphilin-3A and positively regulates AVP secretion. These data shed light on a novel potential mechanism involving rabphilin-3A in AVP secretion, and suggest a new role of CAND1 as a regulator of hormone or neurotransmitter secretion.
Collapse
Affiliation(s)
- Kohtaro Nakashima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Seiji Takeuchi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Atsushi Kiyota
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yoshinori Yasuda
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Naoko Iwata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yoshihisa Sugimura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Fujita Health University, Toyoake, Aichi 470-1192 Japan
| |
Collapse
|
26
|
Lozić M, Šarenac O, Murphy D, Japundžić-Žigon N. Vasopressin, Central Autonomic Control and Blood Pressure Regulation. Curr Hypertens Rep 2018; 20:11. [DOI: 10.1007/s11906-018-0811-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
27
|
Althammer F, Grinevich V. Diversity of oxytocin neurons: beyond magno- and parvocellular cell types? J Neuroendocrinol 2017; 30. [PMID: 29024187 DOI: 10.1111/jne.12549] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/09/2017] [Indexed: 01/31/2023]
Abstract
The hypothalamic neuropeptide oxytocin (OT), which is evolutionarily conserved among different species throughout the animal kingdom, is a key modulator of a variety of socio-emotional behaviors such as fear, trust and empathy. OT cells in the mammalian hypothalamus have been traditionally divided into two distinct types - magnocellular (magnOT) and parvocellular (parvOT) or preautonomic neurons. This distinction is based on OT cell sizes and shapes, projections, electrophysiological activity and functions. Indeed, while neuroendocrine magnOT neurons are known to primarily project their axons to the posterior pituitary and to a number of forebrain regions, non-neuroendocrine parvOT neurons have been seen as the main source of OT innervation of the brainstem and spinal cord to control autonomic functions and pain perception. However, very recent findings demonstrated distinct genetic profiles in OT neurons, allowing discrimination of at least four types of cells expressing OT. Furthermore, unexpected axonal projections of parvOT neurons to the forebrain and magnOT neurons to the midbrain have been newly reported. In this review, we focus on the detailed analysis of methods of distinction between OT cell types, in- and output sites, morphology as well as on the direct connectivity between OT neurons and its physiological significance. At the end, we propose a hypothesis that the central OT system is composed of more than just two OT cell types, which should be further verified by the application of available genetic and anatomical techniques. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Schaller Research Group on Neuropeptides at German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
| | - Valery Grinevich
- Schaller Research Group on Neuropeptides at German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
- CellNetworks Cluster of Excellence at the, University of Heidelberg, 69120, Heidelberg, Germany
- Central Institute of Mental Health, Mannheim, 68159, Germany
| |
Collapse
|
28
|
Fernández M, Mollinedo-Gajate I, Peñagarikano O. Neural Circuits for Social Cognition: Implications for Autism. Neuroscience 2017; 370:148-162. [PMID: 28729065 DOI: 10.1016/j.neuroscience.2017.07.013] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/01/2017] [Accepted: 07/07/2017] [Indexed: 12/28/2022]
Abstract
Social neuroscience, the study of the neurobiological basis of social behavior, has become a major area of current research in behavioral neuroscience and psychiatry, since many psychiatric disorders are characterized by social deficits. Social behavior refers to the behavioral response with regard to socially relevant information, and requires the perception and integration of social cues through a complex cognition process (i.e. social cognition) that involves attention, memory, motivation and emotion. Neurobiological and molecular mechanisms underlying social behavior are highly conserved across species, and inter- and intra-specific variability observed in social behavior can be explained to large extent by differential activity of this conserved neural network. Human functional magnetic resonance imaging (fMRI) studies have greatly informed about the brain structures and their connectivity networks that are important for social cognition. Animal research has been crucial for identifying specific circuits and molecular mechanisms that modulate this structural network. From a molecular neurobiology perspective, activity in these brain structures is coordinated by neuronal circuits modulated by several neurotransmitters and neuromodulators. Thus, quantitative variation in the levels, release and/or receptor density of these molecules could affect the observed behavioral response. The present review presents an overall framework of the components of the social brain circuitry and its modulation. By integrating multiple research approaches, from human fMRI studies to animal models we can start shedding light into how dysfunction in these circuits could lead to disorders of social-functioning such as Autism.
Collapse
Affiliation(s)
- Marta Fernández
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Irene Mollinedo-Gajate
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Leioa 48940, Spain; Biomedical Research Networking Center in Mental Health (CIBERSAM), Spain
| | - Olga Peñagarikano
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Leioa 48940, Spain; Biomedical Research Networking Center in Mental Health (CIBERSAM), Spain.
| |
Collapse
|
29
|
Leng G, Sabatier N. Oxytocin - The Sweet Hormone? Trends Endocrinol Metab 2017; 28:365-376. [PMID: 28283319 DOI: 10.1016/j.tem.2017.02.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/07/2017] [Accepted: 02/10/2017] [Indexed: 12/14/2022]
Abstract
Mammalian neurons that produce oxytocin and vasopressin apparently evolved from an ancient cell type with both sensory and neurosecretory properties that probably linked reproductive functions to energy status and feeding behavior. Oxytocin in modern mammals is an autocrine/paracrine regulator of cell function, a systemic hormone, a neuromodulator released from axon terminals within the brain, and a 'neurohormone' that acts at receptors distant from its site of release. In the periphery oxytocin is involved in electrolyte homeostasis, gastric motility, glucose homeostasis, adipogenesis, and osteogenesis, and within the brain it is involved in food reward, food choice, and satiety. Oxytocin preferentially suppresses intake of sweet-tasting carbohydrates while improving glucose tolerance and supporting bone remodeling, making it an enticing translational target.
Collapse
Affiliation(s)
- Gareth Leng
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK.
| | - Nancy Sabatier
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| |
Collapse
|
30
|
Peñagarikano O. Oxytocin in animal models of autism spectrum disorder. Dev Neurobiol 2017; 77:202-213. [DOI: 10.1002/dneu.22449] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/30/2016] [Accepted: 09/04/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Olga Peñagarikano
- Department of Pharmacology, School of Medicine; University of the Basque Country; Sarriena s/n Leioa 48940 Spain
- Center for Biomedical Research Network in Mental Health (CIBERSAM), ISCIII; Spain
| |
Collapse
|
31
|
Constantin S. Progress and Challenges in the Search for the Mechanisms of Pulsatile Gonadotropin-Releasing Hormone Secretion. Front Endocrinol (Lausanne) 2017; 8:180. [PMID: 28790978 PMCID: PMC5523686 DOI: 10.3389/fendo.2017.00180] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/10/2017] [Indexed: 12/05/2022] Open
Abstract
Fertility relies on the proper functioning of the hypothalamic-pituitary-gonadal axis. The hormonal cascade begins with hypothalamic neurons secreting gonadotropin-releasing hormone (GnRH) into the hypophyseal portal system. In turn, the GnRH-activated gonadotrophs in the anterior pituitary release gonadotropins, which then act on the gonads to regulate gametogenesis and sex steroidogenesis. Finally, sex steroids close this axis by feeding back to the hypothalamus. Despite this seeming straightforwardness, the axis is orchestrated by a complex neuronal network in the central nervous system. For reproductive success, GnRH neurons, the final output of this network, must integrate and translate a wide range of cues, both environmental and physiological, to the gonadotrophs via pulsatile GnRH secretion. This secretory profile is critical for gonadotropic function, yet the mechanisms underlying these pulses remain unknown. Literature supports both intrinsically and extrinsically driven GnRH neuronal activity. However, the caveat of the techniques supporting either one of the two hypotheses is the gap between events recorded at a single-cell level and GnRH secretion measured at the population level. This review aims to compile data about GnRH neuronal activity focusing on the physiological output, GnRH secretion.
Collapse
Affiliation(s)
- Stephanie Constantin
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Stephanie Constantin,
| |
Collapse
|
32
|
Ludwig M, Apps D, Menzies J, Patel JC, Rice ME. Dendritic Release of Neurotransmitters. Compr Physiol 2016; 7:235-252. [PMID: 28135005 DOI: 10.1002/cphy.c160007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Release of neuroactive substances by exocytosis from dendrites is surprisingly widespread and is not confined to a particular class of transmitters: it occurs in multiple brain regions, and includes a range of neuropeptides, classical neurotransmitters, and signaling molecules, such as nitric oxide, carbon monoxide, ATP, and arachidonic acid. This review is focused on hypothalamic neuroendocrine cells that release vasopressin and oxytocin and midbrain neurons that release dopamine. For these two model systems, the stimuli, mechanisms, and physiological functions of dendritic release have been explored in greater detail than is yet available for other neurons and neuroactive substances. © 2017 American Physiological Society. Compr Physiol 7:235-252, 2017.
Collapse
Affiliation(s)
- Mike Ludwig
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - David Apps
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - John Menzies
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Jyoti C Patel
- Department of Neurosurgery, New York University School of Medicine, New York, USA
| | - Margaret E Rice
- Department of Neurosurgery, New York University School of Medicine, New York, USA.,Department of Neuroscience and Physiology, New York University School of Medicine, New York, USA
| |
Collapse
|
33
|
Takeuchi S, Iwama S, Takagi H, Kiyota A, Nakashima K, Izumida H, Fujisawa H, Iwata N, Suga H, Watanabe T, Kaibuchi K, Oiso Y, Arima H, Sugimura Y. Tomosyn Negatively Regulates Arginine Vasopressin Secretion in Embryonic Stem Cell-Derived Neurons. PLoS One 2016; 11:e0164544. [PMID: 27732637 PMCID: PMC5061411 DOI: 10.1371/journal.pone.0164544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/27/2016] [Indexed: 11/24/2022] Open
Abstract
Arginine vasopressin (AVP) is secreted via exocytosis; however, the precise molecular mechanism underlying the exocytosis of AVP remains to be elucidated. To better understand the mechanisms of AVP secretion, in our study we have identified proteins that bind with a 25 kDa synaptosomal-associated protein (SNAP25). SNAP25 plays a crucial role in exocytosis, in the posterior pituitary. Embryonic stem (ES) cell-derived AVP neurons were established to investigate the functions of the identified proteins. Using glutathione S-transferase (GST)-pulldown assays and proteomic analyses, we identified tomosyn-1 (syntaxin-binding protein 5) as a SNAP25-binding protein in the posterior pituitary. Coimmunoprecipitation assays indicated that tomosyn formed N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes with SNAP25 and syntaxin1. Immunohistochemistry showed that tomosyn localized to the posterior pituitary. Mouse ES cells self-differentiated into AVP neurons (mES-AVP) that expressed tomosyn and two transmembrane SNARE proteins, including SNAP25 and syntaxin1. KCl increased AVP secretion in mES-AVP, and overexpression of tomosyn-1 reduced KCl-stimulated AVP secretion. Downregulation of tomosyn-1 with siRNA increased KCl-stimulated AVP secretion. These results suggested that tomosyn-1 negatively regulated AVP secretion in mES-AVP and further suggest the possibility of using mES-AVP culture systems to evaluate the role of synaptic proteins from AVP neurons.
Collapse
Affiliation(s)
- Seiji Takeuchi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Kiyota
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kohtaro Nakashima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hisakazu Izumida
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Haruki Fujisawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoko Iwata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Watanabe
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yutaka Oiso
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Sugimura
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail:
| |
Collapse
|
34
|
Van Cappellen P, Way BM, Isgett SF, Fredrickson BL. Effects of oxytocin administration on spirituality and emotional responses to meditation. Soc Cogn Affect Neurosci 2016; 11:1579-87. [PMID: 27317929 DOI: 10.1093/scan/nsw078] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 05/31/2016] [Indexed: 11/12/2022] Open
Abstract
The oxytocin (OT) system, critically involved in social bonding, may also impinge on spirituality, which is the belief in a meaningful life imbued with a sense of connection to a Higher Power and/or the world. Midlife male participants (N = 83) were randomly assigned to receive intranasal OT or placebo. In exploratory analyses, participants were also genotyped for polymorphisms in two genes critical for OT signaling, the oxytocin receptor gene (OXTR rs53576) and CD38 (rs6449182 and rs3796863). Results showed that intranasal OT increased self-reported spirituality on two separate measures and this effect remained significant a week later. It also boosted participants' experience of specific positive emotions during meditation, at both explicit and implicit levels. Furthermore, the effect of OT on spirituality was moderated by OT-related genotypes. These results provide the first experimental evidence that spirituality, endorsed by millions worldwide, appears to be supported by OT.
Collapse
Affiliation(s)
- Patty Van Cappellen
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Baldwin M Way
- Department of Psychology, The Ohio State University, Columbus Ohio, OH, USA
| | - Suzannah F Isgett
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| | - Barbara L Fredrickson
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
35
|
Ludwig M, Stern J. Multiple signalling modalities mediated by dendritic exocytosis of oxytocin and vasopressin. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0182. [PMID: 26009761 DOI: 10.1098/rstb.2014.0182] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The mammalian hypothalamic magnocellular neurons of the supraoptic and paraventricular nuclei are among the best understood of all peptidergic neurons. Through their anatomical features, vasopressin- and oxytocin-containing neurons have revealed many important aspects of dendritic functions. Here, we review our understanding of the mechanisms of somato-dendritic peptide release, and the effects of autocrine, paracrine and hormone-like signalling on neuronal networks and behaviour.
Collapse
Affiliation(s)
- Mike Ludwig
- Centre for Integrative Physiology, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - Javier Stern
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
36
|
Otero-García M, Agustín-Pavón C, Lanuza E, Martínez-García F. Distribution of oxytocin and co-localization with arginine vasopressin in the brain of mice. Brain Struct Funct 2015; 221:3445-73. [PMID: 26388166 DOI: 10.1007/s00429-015-1111-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/09/2015] [Indexed: 12/19/2022]
Abstract
Oxytocin (OT) and vasopressin (AVP) play a major role in social behaviours. Mice have become the species of choice for neurobiology of social behaviour due to identification of mouse pheromones and the advantage of genetically modified mice. However, neuroanatomical data on nonapeptidergic systems in mice are fragmentary, especially concerning the central distribution of OT. Therefore, we analyse the immunoreactivity for OT and its neurophysin in the brain of male and female mice (strain CD1). Further, we combine immunofluorescent detection of OT and AVP to locate cells co-expressing both peptides and their putative axonal processes. The results indicate that OT is present in cells of the neurosecretory paraventricular (Pa) and supraoptic hypothalamic nuclei (SON). From the anterior SON, OTergic cells extend into the medial amygdala, where a sparse cell population occupies its ventral anterior and posterior divisions. Co-expression of OT and AVP in these nuclei is rare. Moreover, a remarkable OTergic cell group is found near the ventral bed nucleus of the stria terminalis (BST), distributed between the anterodorsal preoptic nucleus and the nucleus of anterior commissure (ADP/AC). This cell group, the rostral edge of the Pa and the periventricular hypothalamus display frequent OT + AVP double labelling, with a general dominance of OT over AVP immunoreactivity. Fibres with similar immunoreactivity profile innervate the accumbens shell and core, central amygdala and portions of the intervening BST. These data, together with data in the literature on rats, suggest that the projections of ADP/AC nonapeptidergic cells onto these brain centres could promote pup-motivated behaviours and inhibit pup avoidance during motherhood.
Collapse
Affiliation(s)
- Marcos Otero-García
- Departaments de Biologia Cel·lular i de Biologia Funcional, Facultat de Ciències Biològiques, Universitat de València, València, Spain
| | - Carmen Agustín-Pavón
- Lab. of Functional Neuroanatomy (NeuroFun), Unitat Predepartamental de Medicina, Facultat de Ciències de la Salut, Universitat Jaume I, Av. de Vicent Sos Baynat, s/n, 12071, Castelló de la Plana, Spain
| | - Enrique Lanuza
- Departaments de Biologia Cel·lular i de Biologia Funcional, Facultat de Ciències Biològiques, Universitat de València, València, Spain
| | - Fernando Martínez-García
- Lab. of Functional Neuroanatomy (NeuroFun), Unitat Predepartamental de Medicina, Facultat de Ciències de la Salut, Universitat Jaume I, Av. de Vicent Sos Baynat, s/n, 12071, Castelló de la Plana, Spain.
| |
Collapse
|
37
|
Sladek CD, Michelini LC, Stachenfeld NS, Stern JE, Urban JH. Endocrine‐Autonomic Linkages. Compr Physiol 2015; 5:1281-323. [DOI: 10.1002/cphy.c140028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
38
|
de Souza Mecawi A, Ruginsk SG, Elias LLK, Varanda WA, Antunes‐Rodrigues J. Neuroendocrine Regulation of Hydromineral Homeostasis. Compr Physiol 2015; 5:1465-516. [DOI: 10.1002/cphy.c140031] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
39
|
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: 3.0] [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
|
40
|
Romanov RA, Alpár A, Zhang MD, Zeisel A, Calas A, Landry M, Fuszard M, Shirran SL, Schnell R, Dobolyi Á, Oláh M, Spence L, Mulder J, Martens H, Palkovits M, Uhlen M, Sitte HH, Botting CH, Wagner L, Linnarsson S, Hökfelt T, Harkany T. A secretagogin locus of the mammalian hypothalamus controls stress hormone release. EMBO J 2015; 34:36-54. [PMID: 25430741 PMCID: PMC4291479 DOI: 10.15252/embj.201488977] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 10/07/2014] [Accepted: 10/21/2014] [Indexed: 11/09/2022] Open
Abstract
A hierarchical hormonal cascade along the hypothalamic-pituitary-adrenal axis orchestrates bodily responses to stress. Although corticotropin-releasing hormone (CRH), produced by parvocellular neurons of the hypothalamic paraventricular nucleus (PVN) and released into the portal circulation at the median eminence, is known to prime downstream hormone release, the molecular mechanism regulating phasic CRH release remains poorly understood. Here, we find a cohort of parvocellular cells interspersed with magnocellular PVN neurons expressing secretagogin. Single-cell transcriptome analysis combined with protein interactome profiling identifies secretagogin neurons as a distinct CRH-releasing neuron population reliant on secretagogin's Ca(2+) sensor properties and protein interactions with the vesicular traffic and exocytosis release machineries to liberate this key hypothalamic releasing hormone. Pharmacological tools combined with RNA interference demonstrate that secretagogin's loss of function occludes adrenocorticotropic hormone release from the pituitary and lowers peripheral corticosterone levels in response to acute stress. Cumulatively, these data define a novel secretagogin neuronal locus and molecular axis underpinning stress responsiveness.
Collapse
Affiliation(s)
- Roman A Romanov
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Alán Alpár
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ming-Dong Zhang
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Amit Zeisel
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - André Calas
- Laboratory for Central Mechanisms of Pain Sensitization, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 Université Bordeaux 2, Bordeaux, France
| | - Marc Landry
- Laboratory for Central Mechanisms of Pain Sensitization, Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 Université Bordeaux 2, Bordeaux, France
| | - Matthew Fuszard
- School of Chemistry, University of St. Andrews, St. Andrews, UK
| | - Sally L Shirran
- School of Chemistry, University of St. Andrews, St. Andrews, UK
| | - Robert Schnell
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Árpád Dobolyi
- Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Márk Oláh
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Lauren Spence
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | | | - Miklós Palkovits
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, Hungary
| | - Mathias Uhlen
- Science for Life Laboratory, Albanova University Center, Royal Institute of Technology, Stockholm, Sweden
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology Medical University of Vienna, Vienna, Austria
| | | | - Ludwig Wagner
- University Clinic for Internal Medicine III General Hospital Vienna, Vienna, Austria
| | - Sten Linnarsson
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tibor Harkany
- Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Wodowska J, Ciosek J. Galanin and galanin-like peptide modulate vasopressin and oxytocin release in vitro: the role of galanin receptors. Neuropeptides 2014; 48:387-97. [PMID: 25464889 DOI: 10.1016/j.npep.2014.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 09/18/2014] [Accepted: 10/22/2014] [Indexed: 12/17/2022]
Abstract
Galanin (Gal) and galanin-like peptide (GALP) may be involved in the mechanisms of the hypothalamo-neurohypophysial system. The aim of the present in vitro study was to compare the influence of Gal and GALP on vasopressin (AVP) and oxytocin (OT) release from isolated rat neurohypophysis (NH) or hypothalamo-neurohypophysial explants (Hth-NH). The effect of Gal/GALP on AVP/OT secretion was also studied in the presence of galantide, the non-selective galanin receptors antagonist. Gal at concentrations of 10(-10 )M and 10(-8 )M distinctly inhibited basal and K(+)-stimulated AVP release from the NH and Hth-NH explants, whereas Gal exerted a similar action on OT release only during basal incubation. Gal added to the incubation medium in the presence of galantide did not exert any action on the secretion of either neurohormone from NH and Hth-NH explants. GALP (10(-10 )M and 10(-9 )M) induced intensified basal AVP release from the NH and Hth-NH complex as well as the release of potassium-evoked AVP from the Hth-NH. The same effect of GALP has been observed in the presence of galantide. GALP added to basal incubation medium was the reason for stimulated OT release from the NH as well as from the Hth-NH explants. However, under potassium-stimulated conditions, OT release from the NH and Hth-NH complexes has been observed to be distinctly impaired. Galantide did not block this inhibitory effect of GALP on OT secretion. It may be concluded that: (i) Gal as well as GALP modulate AVP and OT release at every level of the hypothalamo-neurohypophysial system; (ii) Gal acts in the rat central nervous system as the inhibitory neuromodulator for AVP and OT release via its galanin receptors; (iii) the stimulatory effect of GALP on AVP and OT release is likely to be mediated via an unidentified specific GALP receptor(s).
Collapse
Affiliation(s)
- Justyna Wodowska
- Department of Neuropeptides Research, Faculty of Health Sciences, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Joanna Ciosek
- Department of Neuropeptides Research, Faculty of Health Sciences, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland.
| |
Collapse
|
42
|
Milutinović-Smiljanić S, Šarenac O, Lozić-Djurić M, Murphy D, Japundžić-Žigon N. Evidence for involvement of central vasopressin V1b and V2 receptors in stress-induced baroreflex desensitization. Br J Pharmacol 2014; 169:900-8. [PMID: 23488898 DOI: 10.1111/bph.12161] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/16/2013] [Accepted: 02/17/2013] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE It is well recognized that vasopressin modulates the neurogenic control of the circulation. Here, we report the central mechanisms by which vasopressin modulates cardiovascular response to stress induced by immobilization. EXPERIMENTAL APPROACH Experiments were performed in conscious male Wistar rats equipped with radiotelemetric device for continuous measurement of haemodynamic parameters: systolic and diastolic BP and heart rate (HR). The functioning of the spontaneous baro-receptor reflex (BRR) was evaluated using the sequence method and the following parameters were evaluated: BRR sensitivity (BRS) and BRR effectiveness index (BEI). KEY RESULTS Under baseline physiological conditions intracerebroventricular injection of 100 and 500 ng of selective non-peptide V1a or V1b or V2 receptor antagonist did not modify BP, HR and BRR. Rats exposed to 15 min long stress by immobilization exhibited increase of BP, HR, reduction of BRS and no change in BEI. Pretreatment of rats with V1a receptor antagonist did not modulate the BP, HR, BRS and BEI response to stress. Pretreatment of rats with V1b receptor and V2 receptor antagonist, at both doses, prevented BRR desensitization and tachycardia, but failed to modulate stress-induced hypertension. CONCLUSIONS AND IMPLICATIONS Vasopressin by the stimulation of central V1b- and V2-like receptors mediates stress-induced tachycardia and BRR desensitization. If these mechanisms are involved, BRR desensitization in heart failure and hypertension associated with poor outcome, they could be considered as novel targets for cardiovascular drug development.
Collapse
|
43
|
Leon-Pinzon C, Cercós MG, Noguez P, Trueta C, De-Miguel FF. Exocytosis of serotonin from the neuronal soma is sustained by a serotonin and calcium-dependent feedback loop. Front Cell Neurosci 2014; 8:169. [PMID: 25018697 PMCID: PMC4072984 DOI: 10.3389/fncel.2014.00169] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/02/2014] [Indexed: 11/25/2022] Open
Abstract
The soma of many neurons releases large amounts of transmitter molecules through an exocytosis process that continues for hundreds of seconds after the end of the triggering stimulus. Transmitters released in this way modulate the activity of neurons, glia and blood vessels over vast volumes of the nervous system. Here we studied how somatic exocytosis is maintained for such long periods in the absence of electrical stimulation and transmembrane Ca(2+) entry. Somatic exocytosis of serotonin from dense core vesicles could be triggered by a train of 10 action potentials at 20 Hz in Retzius neurons of the leech. However, the same number of action potentials produced at 1 Hz failed to evoke any exocytosis. The 20-Hz train evoked exocytosis through a sequence of intracellular Ca(2+) transients, with each transient having a different origin, timing and intracellular distribution. Upon electrical stimulation, transmembrane Ca(2+) entry through L-type channels activated Ca(2+)-induced Ca(2+) release. A resulting fast Ca(2+) transient evoked an early exocytosis of serotonin from sparse vesicles resting close to the plasma membrane. This Ca(2+) transient also triggered the transport of distant clusters of vesicles toward the plasma membrane. Upon exocytosis, the released serotonin activated autoreceptors coupled to phospholipase C, which in turn produced an intracellular Ca(2+) increase in the submembrane shell. This localized Ca(2+) increase evoked new exocytosis as the vesicles in the clusters arrived gradually at the plasma membrane. In this way, the extracellular serotonin elevated the intracellular Ca(2+) and this Ca(2+) evoked more exocytosis. The resulting positive feedback loop maintained exocytosis for the following hundreds of seconds until the last vesicles in the clusters fused. Since somatic exocytosis displays similar kinetics in neurons releasing different types of transmitters, the data presented here contributes to understand the cellular basis of paracrine neurotransmission.
Collapse
Affiliation(s)
- Carolina Leon-Pinzon
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico D.F., México
| | - Montserrat G. Cercós
- Departamento de Neurofisiología, Instituto Nacional de Psiquiatriìa Ramoìn de la Fuente MunñizMéxico D.F., México
| | - Paula Noguez
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico D.F., México
| | - Citlali Trueta
- Departamento de Neurofisiología, Instituto Nacional de Psiquiatriìa Ramoìn de la Fuente MunñizMéxico D.F., México
| | - Francisco F. De-Miguel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico D.F., México
| |
Collapse
|
44
|
Chen M, Križaj D, Thoreson WB. Intracellular calcium stores drive slow non-ribbon vesicle release from rod photoreceptors. Front Cell Neurosci 2014; 8:20. [PMID: 24550779 PMCID: PMC3910126 DOI: 10.3389/fncel.2014.00020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/13/2014] [Indexed: 01/26/2023] Open
Abstract
Rods are capable of greater slow release than cones contributing to overall slower release kinetics. Slow release in rods involves Ca2+-induced Ca2+ release (CICR). By impairing release from ribbons, we found that unlike cones where release occurs entirely at ribbon-style active zones, slow release from rods occurs mostly at ectopic, non-ribbon sites. To investigate the role of CICR in ribbon and non-ribbon release from rods, we used total internal reflection fluorescence microscopy as a tool for visualizing terminals of isolated rods loaded with fluorescent Ca2+ indicator dyes and synaptic vesicles loaded with dextran-conjugated pH-sensitive rhodamine. We found that rather than simply facilitating release, activation of CICR by ryanodine triggered release directly in rods, independent of plasma membrane Ca2+ channel activation. Ryanodine-evoked release occurred mostly at non-ribbon sites and release evoked by sustained depolarization at non-ribbon sites was mostly due to CICR. Unlike release at ribbon-style active zones, non-ribbon release did not occur at fixed locations. Fluorescence recovery after photobleaching of endoplasmic reticulum (ER)-tracker dye in rod terminals showed that ER extends continuously from synapse to soma. Release of Ca2+ from terminal ER by lengthy depolarization did not significantly deplete Ca2+ from ER in the perikaryon. Collectively, these results indicate that CICR-triggered release at non-ribbon sites is a major mechanism for maintaining vesicle release from rods and that CICR in terminals may be sustained by diffusion of Ca2+ through ER from other parts of the cell.
Collapse
Affiliation(s)
- Minghui Chen
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center Omaha, NE, USA ; Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center Omaha, NE, USA
| | - David Križaj
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah School of Medicine Salt Lake City, UT, USA
| | - Wallace B Thoreson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center Omaha, NE, USA ; Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center Omaha, NE, USA
| |
Collapse
|
45
|
Faje AT, Nachtigall L, Wexler D, Miller KK, Klibanski A, Makimura H. Central diabetes insipidus: a previously unreported side effect of temozolomide. J Clin Endocrinol Metab 2013; 98:3926-31. [PMID: 23928668 PMCID: PMC3790614 DOI: 10.1210/jc.2013-2435] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Temozolomide (TMZ) is an alkylating agent primarily used to treat tumors of the central nervous system. We describe 2 patients with apparent TMZ-induced central diabetes insipidus. Using our institution's Research Patient Database Registry, we identified 3 additional potential cases of TMZ-induced diabetes insipidus among a group of 1545 patients treated with TMZ. CASE PRESENTATIONS A 53-year-old male with an oligoastrocytoma and a 38-year-old male with an oligodendroglioma each developed symptoms of polydipsia and polyuria approximately 2 months after the initiation of TMZ. Laboratory analyses demonstrated hypernatremia and urinary concentrating defects, consistent with the presence of diabetes insipidus, and the patients were successfully treated with desmopressin acetate. Desmopressin acetate was withdrawn after the discontinuation of TMZ, and diabetes insipidus did not recur. Magnetic resonance imaging of the pituitary and hypothalamus was unremarkable apart from the absence of a posterior pituitary bright spot in both of the cases. Anterior pituitary function tests were normal in both cases. Using the Research Patient Database Registry database, we identified the 2 index cases and 3 additional potential cases of diabetes insipidus for an estimated prevalence of 0.3% (5 cases of diabetes insipidus per 1545 patients prescribed TMZ). CONCLUSIONS Central diabetes insipidus is a rare but reversible side effect of treatment with TMZ.
Collapse
Affiliation(s)
- Alexander T Faje
- MD, BUL 457, Neuroendocrine Unit, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114.
| | | | | | | | | | | |
Collapse
|
46
|
Singh S, More KR, Chitnis CE. Role of calcineurin and actin dynamics in regulated secretion of microneme proteins in Plasmodium falciparum merozoites during erythrocyte invasion. Cell Microbiol 2013; 16:50-63. [PMID: 23910910 DOI: 10.1111/cmi.12177] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 07/16/2013] [Accepted: 07/30/2013] [Indexed: 11/28/2022]
Abstract
Plasmodium falciparum invades host erythrocytes by multiple invasion pathways. The invasion of erythrocytes by P. falciparum merozoites is a complex process that requires multiple interactions between host receptors and parasite ligands. A number of parasite proteins that mediate interaction with host receptors during invasion are localized to membrane-bound apical organelles referred to as micronemes and rhoptries. The timely release of these proteins to the merozoite surface is crucial for receptor engagement and invasion. It has been demonstrated previously that exposure of merozoites to a low potassium (K(+)) ionic environment as found in blood plasma leads to a rise in cytosolic calcium (Ca(2+)), which triggers microneme secretion. The signalling pathways that regulate microneme discharge in response to rise in cytosolic Ca(2+) are not completely understood. Here, we show that a P. falciparum Ca(2+)-dependent protein phosphatase, calcineurin (PfCN), is an essential regulator of Ca(2+)-dependent microneme exocytosis. An increase in PfCN activity was observed in merozoites following exposure to a low K(+) environment. Treatment of merozoites with calcineurin inhibitors such as FK506 and cyclosporin A prior to transfer to a low K(+) environment resulted in inhibition of secretion of microneme protein apical merozoite antigen-1 (PfAMA-1). Inhibition of PfCN was shown to result in reduced dephosphorylation and depolymerization of apical actin, which appears to be criticalfor microneme secretion. PfCN thus serves as an effector of Ca(2+)-dependent microneme exocytosis by regulating depolymerization of apical actin. Inhibitors that target PfCN block microneme exocytosis and limit growth of P. falciparum blood-stage parasites providing a novel approach towards development of new therapeutic strategies against malaria.
Collapse
Affiliation(s)
- Shailja Singh
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | | | | |
Collapse
|
47
|
Trueta C, De-Miguel FF. Extrasynaptic exocytosis and its mechanisms: a source of molecules mediating volume transmission in the nervous system. Front Physiol 2012; 3:319. [PMID: 22969726 PMCID: PMC3432928 DOI: 10.3389/fphys.2012.00319] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 07/21/2012] [Indexed: 11/14/2022] Open
Abstract
We review the evidence of exocytosis from extrasynaptic sites in the soma, dendrites, and axonal varicosities of central and peripheral neurons of vertebrates and invertebrates, with emphasis on somatic exocytosis, and how it contributes to signaling in the nervous system. The finding of secretory vesicles in extrasynaptic sites of neurons, the presence of signaling molecules (namely transmitters or peptides) in the extracellular space outside synaptic clefts, and the mismatch between exocytosis sites and the location of receptors for these molecules in neurons and glial cells, have long suggested that in addition to synaptic communication, transmitters are released, and act extrasynaptically. The catalog of these molecules includes low molecular weight transmitters such as monoamines, acetylcholine, glutamate, gama-aminobutiric acid (GABA), adenosine-5-triphosphate (ATP), and a list of peptides including substance P, brain-derived neurotrophic factor (BDNF), and oxytocin. By comparing the mechanisms of extrasynaptic exocytosis of different signaling molecules by various neuron types we show that it is a widespread mechanism for communication in the nervous system that uses certain common mechanisms, which are different from those of synaptic exocytosis but similar to those of exocytosis from excitable endocrine cells. Somatic exocytosis has been measured directly in different neuron types. It starts after high-frequency electrical activity or long experimental depolarizations and may continue for several minutes after the end of stimulation. Activation of L-type calcium channels, calcium release from intracellular stores and vesicle transport towards the plasma membrane couple excitation and exocytosis from small clear or large dense core vesicles in release sites lacking postsynaptic counterparts. The presence of synaptic and extrasynaptic exocytosis endows individual neurons with a wide variety of time- and space-dependent communication possibilities. Extrasynaptic exocytosis may be the major source of signaling molecules producing volume transmission and by doing so may be part of a long duration signaling mode in the nervous system.
Collapse
Affiliation(s)
- Citlali Trueta
- Departamento de Neurofisiología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz México, D.F., México
| | | |
Collapse
|