1
|
Jung JW, Kim YJ, Choi JS, Goto Y, Lee YA. Dopamine and serotonin alterations by Hizikia fusiformis extracts under in vitro cortical primary neuronal cell cultures. Nutr Res Pract 2023; 17:408-420. [PMID: 37266125 PMCID: PMC10232209 DOI: 10.4162/nrp.2023.17.3.408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/23/2022] [Accepted: 01/05/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND/OBJECTIVES Hizikia fusiformis (HF) is a class of brown seaweeds whose active ingredients exert central nervous system protective effects, such as neuroprotection; however, the underlying mechanisms remain unknown. Given that dopamine (DA) and serotonin (5HT) are two major neurotransmitters involved in various psychiatric disorders and neuronal growth in early neurodevelopmental processes, we investigated whether HF extract could modulate the molecular expression associated with DA and 5HT transmission as well as the structural formation of neurons. MATERIALS/METHODS In vitro cell cultures were prepared from cerebral cortical neurons obtained from CD-1 mice on embryonic day 14. Cultured cells were treated with 0.1, 1.0, or 10.0 μg/mL of HT extract for 24 h, followed by fluorescence immunostaining for DA and 5HT-related receptors and transporters and some neuronal structural formation-associated molecules. RESULTS HF extract dose-dependently upregulated the expression levels of selective DA and 5HT receptors, and downregulated the levels of DA and 5HT transporters. Moreover, HF extract increased the neurofilament light chain expression. CONCLUSION These results suggest that HF may modulate DA and 5HT transmission, thereby affecting neurodevelopment.
Collapse
Affiliation(s)
- Jae-Won Jung
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan 38430, Korea
| | - Ye-Jin Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan 38430, Korea
| | - Jae Sue Choi
- Department of Food and Life Sciences, Pukyoung National University, Busan 48513, Korea
| | - Yukiori Goto
- Department of Artificial Intelligence and Technology, Graduate School of Informatics, Kyoto University, Kyoto 606-8501, Japan
| | - Young-A Lee
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan 38430, Korea
| |
Collapse
|
2
|
van Rhijn JR, Shi Y, Bormann M, Mossink B, Frega M, Recaioglu H, Hakobjan M, Klein Gunnewiek T, Schoenmaker C, Palmer E, Faivre L, Kittel-Schneider S, Schubert D, Brunner H, Franke B, Nadif Kasri N. Brunner syndrome associated MAOA mutations result in NMDAR hyperfunction and increased network activity in human dopaminergic neurons. Neurobiol Dis 2021; 163:105587. [PMID: 34923109 DOI: 10.1016/j.nbd.2021.105587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 01/15/2023] Open
Abstract
Monoamine neurotransmitter abundance affects motor control, emotion, and cognitive function and is regulated by monoamine oxidases. Among these, Monoamine oxidase A (MAOA) catalyzes the degradation of dopamine, norepinephrine, and serotonin into their inactive metabolites. Loss-of-function mutations in the X-linked MAOA gene have been associated with Brunner syndrome, which is characterized by various forms of impulsivity, maladaptive externalizing behavior, and mild intellectual disability. Impaired MAOA activity in individuals with Brunner syndrome results in bioamine aberration, but it is currently unknown how this affects neuronal function, specifically in dopaminergic (DA) neurons. Here we generated human induced pluripotent stem cell (hiPSC)-derived DA neurons from three individuals with Brunner syndrome carrying different mutations and characterized neuronal properties at the single cell and neuronal network level in vitro. DA neurons of Brunner syndrome patients showed reduced synaptic density but exhibited hyperactive network activity. Intrinsic functional properties and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-mediated synaptic transmission were not affected in DA neurons of individuals with Brunner syndrome. Instead, we show that the neuronal network hyperactivity is mediated by upregulation of the GRIN2A and GRIN2B subunits of the N-methyl-d-aspartate receptor (NMDAR), resulting in increased NMDAR-mediated currents. By correcting a MAOA missense mutation with CRISPR/Cas9 genome editing we normalized GRIN2A and GRIN2B expression, NMDAR function and neuronal population activity to control levels. Our data suggest that MAOA mutations in Brunner syndrome increase the activity of dopaminergic neurons through upregulation of NMDAR function, which may contribute to the etiology of Brunner syndrome associated phenotypes.
Collapse
Affiliation(s)
- Jon-Ruben van Rhijn
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yan Shi
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Maren Bormann
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Britt Mossink
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Monica Frega
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Clinical neurophysiology, University of Twente, 7522 NB Enschede, Netherlands
| | - Hatice Recaioglu
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marina Hakobjan
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Teun Klein Gunnewiek
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Chantal Schoenmaker
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elizabeth Palmer
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW, Australia; School of Women's and Children's Health, University of New South Wales, Randwick, NSW, Australia
| | - Laurence Faivre
- Centre de Référence Anomalies du développement et Syndromes malformatifs and FHU TRANSLAD, Hôpital d'Enfants, Dijon, France; INSERM UMR1231 GAD, Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe-University, Frankfurt, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Han Brunner
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Clinical Genetics, MUMC+, GROW School of Oncology and Developmental Biology, and MHeNS School of Neuroscience and Maastricht University, Maastricht, the Netherlands
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Psychiatry, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nael Nadif Kasri
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
3
|
An injury-induced serotonergic neuron subpopulation contributes to axon regrowth and function restoration after spinal cord injury in zebrafish. Nat Commun 2021; 12:7093. [PMID: 34876587 PMCID: PMC8651775 DOI: 10.1038/s41467-021-27419-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 11/18/2021] [Indexed: 11/26/2022] Open
Abstract
Spinal cord injury (SCI) interrupts long-projecting descending spinal neurons and disrupts the spinal central pattern generator (CPG) that controls locomotion. The intrinsic mechanisms underlying re-wiring of spinal neural circuits and recovery of locomotion after SCI are unclear. Zebrafish shows axonal regeneration and functional recovery after SCI making it a robust model to study mechanisms of regeneration. Here, we use a two-cut SCI model to investigate whether recovery of locomotion can occur independently of supraspinal connections. Using this injury model, we show that injury induces the localization of a specialized group of intraspinal serotonergic neurons (ISNs), with distinctive molecular and cellular properties, at the injury site. This subpopulation of ISNs have hyperactive terminal varicosities constantly releasing serotonin activating 5-HT1B receptors, resulting in axonal regrowth of spinal interneurons. Axon regrowth of excitatory interneurons is more pronounced compared to inhibitory interneurons. Knock-out of htr1b prevents axon regrowth of spinal excitatory interneurons, negatively affecting coordination of rostral-caudal body movements and restoration of locomotor function. On the other hand, treatment with 5-HT1B receptor agonizts promotes functional recovery following SCI. In summary, our data show an intraspinal mechanism where a subpopulation of ISNs stimulates axonal regrowth resulting in improved recovery of locomotor functions following SCI in zebrafish.
Collapse
|
4
|
Vicenzi S, Foa L, Gasperini RJ. Serotonin functions as a bidirectional guidance molecule regulating growth cone motility. Cell Mol Life Sci 2021; 78:2247-2262. [PMID: 32939562 PMCID: PMC11072016 DOI: 10.1007/s00018-020-03628-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/14/2020] [Accepted: 08/21/2020] [Indexed: 02/02/2023]
Abstract
The neurotransmitter serotonin has been implicated in a range of complex neurological disorders linked to alterations of neuronal circuitry. Serotonin is synthesized in the developing brain before most neuronal circuits become fully functional, suggesting that serotonin might play a distinct regulatory role in shaping circuits prior to its function as a classical neurotransmitter. In this study, we asked if serotonin acts as a guidance cue by examining how serotonin alters growth cone motility of rodent sensory neurons in vitro. Using a growth cone motility assay, we found that serotonin acted as both an attractive and repulsive guidance cue through a narrow concentration range. Extracellular gradients of 50 µM serotonin elicited attraction, mediated by the serotonin 5-HT2a receptor while 100 µM serotonin elicited repulsion mediated by the 5-HT1b receptor. Importantly, high resolution imaging of growth cones indicated that these receptors signalled through their canonical pathways of endoplasmic reticulum-mediated calcium release and cAMP depletion, respectively. This novel characterisation of growth cone motility in response to serotonin gradients provides compelling evidence that secreted serotonin acts at the molecular level as an axon guidance cue to shape neuronal circuit formation during development.
Collapse
Affiliation(s)
- Silvia Vicenzi
- School of Medicine, University of Tasmania, Hobart, Australia
| | - Lisa Foa
- School of Psychological Sciences, University of Tasmania, Hobart, Australia
| | | |
Collapse
|
5
|
Vahid-Ansari F, Albert PR. Rewiring of the Serotonin System in Major Depression. Front Psychiatry 2021; 12:802581. [PMID: 34975594 PMCID: PMC8716791 DOI: 10.3389/fpsyt.2021.802581] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Serotonin is a key neurotransmitter that is implicated in a wide variety of behavioral and cognitive phenotypes. Originating in the raphe nuclei, 5-HT neurons project widely to innervate many brain regions implicated in the functions. During the development of the brain, as serotonin axons project and innervate brain regions, there is evidence that 5-HT plays key roles in wiring the developing brain, both by modulating 5-HT innervation and by influencing synaptic organization within corticolimbic structures. These actions are mediated by 14 different 5-HT receptors, with region- and cell-specific patterns of expression. More recently, the role of the 5-HT system in synaptic re-organization during adulthood has been suggested. The 5-HT neurons have the unusual capacity to regrow and reinnervate brain regions following insults such as brain injury, chronic stress, or altered development that result in disconnection of the 5-HT system and often cause depression, anxiety, and cognitive impairment. Chronic treatment with antidepressants that amplify 5-HT action, such as selective serotonin reuptake inhibitors (SSRIs), appears to accelerate the rewiring of the 5-HT system by mechanisms that may be critical to the behavioral and cognitive improvements induced in these models. In this review, we survey the possible 5-HT receptor mechanisms that could mediate 5-HT rewiring and assess the evidence that 5-HT-mediated brain rewiring is impacting recovery from mental illness. By amplifying 5-HT-induced rewiring processes using SSRIs and selective 5-HT agonists, more rapid and effective treatments for injury-induced mental illness or cognitive impairment may be achieved.
Collapse
Affiliation(s)
- Faranak Vahid-Ansari
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
6
|
Hanswijk SI, Spoelder M, Shan L, Verheij MMM, Muilwijk OG, Li W, Liu C, Kolk SM, Homberg JR. Gestational Factors throughout Fetal Neurodevelopment: The Serotonin Link. Int J Mol Sci 2020; 21:E5850. [PMID: 32824000 PMCID: PMC7461571 DOI: 10.3390/ijms21165850] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
Serotonin (5-HT) is a critical player in brain development and neuropsychiatric disorders. Fetal 5-HT levels can be influenced by several gestational factors, such as maternal genotype, diet, stress, medication, and immune activation. In this review, addressing both human and animal studies, we discuss how these gestational factors affect placental and fetal brain 5-HT levels, leading to changes in brain structure and function and behavior. We conclude that gestational factors are able to interact and thereby amplify or counteract each other's impact on the fetal 5-HT-ergic system. We, therefore, argue that beyond the understanding of how single gestational factors affect 5-HT-ergic brain development and behavior in offspring, it is critical to elucidate the consequences of interacting factors. Moreover, we describe how each gestational factor is able to alter the 5-HT-ergic influence on the thalamocortical- and prefrontal-limbic circuitry and the hypothalamo-pituitary-adrenocortical-axis. These alterations have been associated with risks to develop attention deficit hyperactivity disorder, autism spectrum disorders, depression, and/or anxiety. Consequently, the manipulation of gestational factors may be used to combat pregnancy-related risks for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sabrina I. Hanswijk
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Marcia Spoelder
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Ling Shan
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands;
| | - Michel M. M. Verheij
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Otto G. Muilwijk
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| | - Weizhuo Li
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China; (W.L.); (C.L.)
| | - Chunqing Liu
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China; (W.L.); (C.L.)
| | - Sharon M. Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 AJ Nijmegen, The Netherlands;
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen Medical Centre, 6525 EN Nijmegen, The Netherlands; (S.I.H.); (M.S.); (M.M.M.V.); (O.G.M.)
| |
Collapse
|
7
|
Vargas J, Alfaro-Rodríguez A, Perez-Orive J. Serotonin induces or inhibits neuritic regeneration of leech CNS neurons depending on neuronal identity. ACTA ACUST UNITED AC 2019; 52:e7988. [PMID: 30785479 PMCID: PMC6376320 DOI: 10.1590/1414-431x20187988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/05/2018] [Indexed: 11/25/2022]
Abstract
Recovery of motor function after central nervous system (CNS) injury is dependent on the regeneration capacity of the nervous system, which is a multifactorial process influenced, among other things, by the role of neuromodulators such as serotonin. The neurotransmitter serotonin can promote neuronal regeneration but there are also reports of it causing restriction, so it is important to clarify these divergent findings in order to understand the direct scope and side effects of potential pharmacological treatments. We evaluated the effect of serotonin on the extent of neuritic outgrowth and morphology of three different neuronal types in the leech Haementeria officinalis during their regeneration in vitro: Retzius interneurons (Rz), annulus erector (AE) motoneurons, and anterolateral number 1 (AL1) CNS neurons. Neurons were isolated and cultured in L15 medium, with or without serotonin. Growth parameters were registered and quantified, and observed differences were analyzed. The addition of serotonin was found to induce AL1 neurons to increase their average growth dramatically by 8.3-fold (P=0.02; n=5), and to have no clear effect on AE motoneurons (P=0.44; n=5). For Rz interneurons, which normally do not regenerate their neurites, the addition of concanavaline-A causes substantial growth, which serotonin was found to inhibit on average by 98% (P=0.02; n=5). The number of primary neurites and their branches were also affected. These results reveal that depending on the neuronal type, serotonin can promote, inhibit, or have no effect on neuronal regeneration. This suggests that after CNS injury, non-specific pharmacological treatments affecting serotonin may have different effects on different neuronal populations.
Collapse
Affiliation(s)
- J Vargas
- Regeneration Laboratory, National Rehabilitation Institute "Luis Guillermo Ibarra Ibarra", Col. Arenal de Guadalupe, Delegacion Tlalpan, Mexico City, Mexico
| | - A Alfaro-Rodríguez
- Neuroscience Division, National Rehabilitation Institute "Luis Guillermo Ibarra Ibarra", Col. Arenal de Guadalupe, Delegacion Tlalpan, Mexico City, Mexico
| | - J Perez-Orive
- Neuroscience Division, National Rehabilitation Institute "Luis Guillermo Ibarra Ibarra", Col. Arenal de Guadalupe, Delegacion Tlalpan, Mexico City, Mexico
| |
Collapse
|
8
|
Roles of 5-HT on phase transition of neurite outgrowth in the identified serotoninergic neuron C1, Helisoma trivolvis. INVERTEBRATE NEUROSCIENCE 2018; 18:10. [PMID: 30128715 DOI: 10.1007/s10158-018-0214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/27/2018] [Indexed: 10/28/2022]
Abstract
Neurite outgrowth is a morphological marker of neuronal differentiation and neuroregeneration, and the process includes four essential phases, namely initiation, elongation, guidance and cessation. Intrinsic and extrinsic signaling molecules seem to involve morphological changes of neurite outgrowth via various cellular signaling cascades phase transition. Although mechanisms associated with neurite outgrowth have been studied extensively, little is known about how phase transition is regulated during neurite outgrowth. 5-HT has long been studied with regard to its relationship to neurite outgrowth in invertebrate and vertebrate culture systems, and many studies have suggested 5-HT inhibits neurite elongation and growth cone motility, in particular, at the growing parts of neurite such as growth cones and filopodia. However, the underlying mechanisms need to be investigated. In this study, we investigated roles of 5-HT on neurite outgrowth using single serotonergic neurons C1 isolated from Helisoma trivolvis. We observed that 5-HT delayed phase transitions from initiation to elongation of neurite outgrowth. This study for the first time demonstrated that 5-HT has a critical role in phase-controlling mechanisms of neurite outgrowth in neuronal cell cultures.
Collapse
|
9
|
Wirth A, Holst K, Ponimaskin E. How serotonin receptors regulate morphogenic signalling in neurons. Prog Neurobiol 2017; 151:35-56. [DOI: 10.1016/j.pneurobio.2016.03.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/09/2016] [Accepted: 03/19/2016] [Indexed: 11/25/2022]
|
10
|
Yu HP, Zhang N, Zhang T, Wang ZL, Li N, Tang HH, Zhang R, Zhang MN, Xu B, Fang Q, Wang R. Activation of NPFF 2 receptor stimulates neurite outgrowth in Neuro 2A cells through activation of ERK signaling pathway. Peptides 2016; 86:24-32. [PMID: 27669639 DOI: 10.1016/j.peptides.2016.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 10/21/2022]
Abstract
Neurite outgrowth is an important process in neural regeneration and plasticity, especially after neural injury, and recent evidence indicates that several Gαi/o protein-coupled receptors play an important role in neurite outgrowth. The neuropeptide (NP)FF system contains two Gαi/o protein-coupled receptors, NPFF1 and NPFF2 receptors, which are mainly distributed in the central nervous system. The aim of the present study was to determine whether the NPFF system is involved in neurite outgrowth in Neuro 2A cells. We showed that Neuro 2A cells endogenously expressed NPFF2 receptor, and the NPFF2 receptor agonist dNPA inhibited cyclic adenosine monophosphate (cAMP) production stimulated by forskolin in Neuro 2A cells. We also demonstrated that NPFF and dNPA dose-dependently induced neurite outgrowth in Neuro 2A cells, which was completely abolished by the NPFF receptor antagonist RF9. Pretreatment with mitogen-activated protein kinase inhibitors PD98059 and U0126 decreased dNPA-induced neurite outgrowth. In addition, dNPA increased phosphorylation of extracellular signal-regulated kinase (ERK) in Neuro 2A cells, which was completely antagonized by pretreatment with U0126. Our results suggest that activation of NPFF2 receptor stimulates neurite outgrowth in Neuro 2A cells through activation of the ERK signaling pathway. Moreover, NPFF2 receptor may be a potential therapeutic target for neural injury and degeneration in the future.
Collapse
Affiliation(s)
- Hong-Ping Yu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Nan Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Ting Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Zi-Long Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Ning Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Hong-Hai Tang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Run Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Meng-Na Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China.
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China.
| |
Collapse
|
11
|
Ishiguro T, Sakata-Haga H, Fukui Y. A 5-HT2A/2C receptor agonist, 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane, mitigates developmental neurotoxicity of ethanol to serotonergic neurons. Congenit Anom (Kyoto) 2016; 56:163-71. [PMID: 26714672 DOI: 10.1111/cga.12152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/10/2015] [Accepted: 12/23/2015] [Indexed: 11/27/2022]
Abstract
Prenatal ethanol exposure causes the reduction of serotonergic (5-HTergic) neurons in the midbrain raphe nuclei. In the present study, we examined whether an activation of signaling via 5-HT2A and 5-HT2C receptors during the fetal period is able to prevent the reduction of 5-HTergic neurons induced by prenatal ethanol exposure. Pregnant Sprague-Dawley rats were given a liquid diet containing 2.5 to 5.0% (w/v) ethanol on gestational days (GDs) 10 to 20 (Et). As a pair-fed control, other pregnant rats were fed the same liquid diet except that the ethanol was replaced by isocaloric sucrose (Pf). Each Et and Pf group was subdivided into two groups; one of the groups was treated with 1 mg/kg (i.p.) of 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI), an agonist for 5-HT2A/2C receptors, during GDs 13 to 19 (Et-DOI or Pf-DOI), and another was injected with saline vehicle only (Et-Sal or Pf-Sal). Their fetuses were removed by cesarean section on GD 19 or 20, and fetal brains were collected. An immunohistological examination of 5-HTergic neurons in the fetuses on embryonic day 20 using an antibody against tryptophan hydroxylase revealed that the number of 5-HTergic neurons in the midbrain raphe nuclei was significantly reduced in the Et-Sal fetuses compared to that of the Pf-Sal and Pf-DOI fetuses, whereas there were no significant differences between Et-DOI and each Pf control. Thus, we concluded that the reduction of 5-HTergic neurons that resulted in prenatal ethanol exposure could be alleviated by the enhancement of signaling via 5-HT2A/2C receptors during the fetal period.
Collapse
Affiliation(s)
- Tsukasa Ishiguro
- Department of Anatomy and Developmental Neurobiology, Tokushima University Graduate School of Biomedical Science, Tokushima, Japan
| | - Hiromi Sakata-Haga
- Department of Anatomy and Developmental Neurobiology, Tokushima University Graduate School of Biomedical Science, Tokushima, Japan
| | - Yoshihiro Fukui
- Department of Anatomy and Developmental Neurobiology, Tokushima University Graduate School of Biomedical Science, Tokushima, Japan
| |
Collapse
|
12
|
Sierra-Fonseca JA, Najera O, Martinez-Jurado J, Walker EM, Varela-Ramirez A, Khan AM, Miranda M, Lamango NS, Roychowdhury S. Nerve growth factor induces neurite outgrowth of PC12 cells by promoting Gβγ-microtubule interaction. BMC Neurosci 2014; 15:132. [PMID: 25552352 PMCID: PMC4302597 DOI: 10.1186/s12868-014-0132-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 11/27/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Assembly and disassembly of microtubules (MTs) is critical for neurite outgrowth and differentiation. Evidence suggests that nerve growth factor (NGF) induces neurite outgrowth from PC12 cells by activating the receptor tyrosine kinase, TrkA. G protein-coupled receptors (GPCRs) as well as heterotrimeric G proteins are also involved in regulating neurite outgrowth. However, the possible connection between these pathways and how they might ultimately converge to regulate the assembly and organization of MTs during neurite outgrowth is not well understood. RESULTS Here, we report that Gβγ, an important component of the GPCR pathway, is critical for NGF-induced neuronal differentiation of PC12 cells. We have found that NGF promoted the interaction of Gβγ with MTs and stimulated MT assembly. While Gβγ-sequestering peptide GRK2i inhibited neurite formation, disrupted MTs, and induced neurite damage, the Gβγ activator mSIRK stimulated neurite outgrowth, which indicates the involvement of Gβγ in this process. Because we have shown earlier that prenylation and subsequent methylation/demethylation of γ subunits are required for the Gβγ-MTs interaction in vitro, small-molecule inhibitors (L-28 and L-23) targeting prenylated methylated protein methyl esterase (PMPMEase) were tested in the current study. We found that these inhibitors disrupted Gβγ and ΜΤ organization and affected cellular morphology and neurite outgrowth. In further support of a role of Gβγ-MT interaction in neuronal differentiation, it was observed that overexpression of Gβγ in PC12 cells induced neurite outgrowth in the absence of added NGF. Moreover, overexpressed Gβγ exhibited a pattern of association with MTs similar to that observed in NGF-differentiated cells. CONCLUSIONS Altogether, our results demonstrate that βγ subunit of heterotrimeric G proteins play a critical role in neurite outgrowth and differentiation by interacting with MTs and modulating MT rearrangement.
Collapse
Affiliation(s)
- Jorge A Sierra-Fonseca
- />Neuromodulation Disorders Cluster, Border Biomedical Research Center, University of Texas, El Paso, TX 79968 USA
- />Department of Biological Sciences, University of Texas, El Paso, TX 79968 USA
- />Present Address: Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Omar Najera
- />Neuromodulation Disorders Cluster, Border Biomedical Research Center, University of Texas, El Paso, TX 79968 USA
- />Department of Biological Sciences, University of Texas, El Paso, TX 79968 USA
| | - Jessica Martinez-Jurado
- />Neuromodulation Disorders Cluster, Border Biomedical Research Center, University of Texas, El Paso, TX 79968 USA
- />Department of Biological Sciences, University of Texas, El Paso, TX 79968 USA
| | - Ellen M Walker
- />Neuromodulation Disorders Cluster, Border Biomedical Research Center, University of Texas, El Paso, TX 79968 USA
- />Department of Biological Sciences, University of Texas, El Paso, TX 79968 USA
| | - Armando Varela-Ramirez
- />Cytometry Screening and Imaging Core facility, Border Biomedical Research Center, University of Texas, El Paso, TX 79968 USA
- />Department of Biological Sciences, University of Texas, El Paso, TX 79968 USA
| | - Arshad M Khan
- />Neuromodulation Disorders Cluster, Border Biomedical Research Center, University of Texas, El Paso, TX 79968 USA
- />Department of Biological Sciences, University of Texas, El Paso, TX 79968 USA
| | - Manuel Miranda
- />Neuromodulation Disorders Cluster, Border Biomedical Research Center, University of Texas, El Paso, TX 79968 USA
- />Department of Biological Sciences, University of Texas, El Paso, TX 79968 USA
| | - Nazarius S Lamango
- />College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307 USA
| | - Sukla Roychowdhury
- />Neuromodulation Disorders Cluster, Border Biomedical Research Center, University of Texas, El Paso, TX 79968 USA
- />Department of Biological Sciences, University of Texas, El Paso, TX 79968 USA
| |
Collapse
|
13
|
Moxon KA, Oliviero A, Aguilar J, Foffani G. Cortical reorganization after spinal cord injury: always for good? Neuroscience 2014; 283:78-94. [PMID: 24997269 DOI: 10.1016/j.neuroscience.2014.06.056] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/09/2014] [Accepted: 06/25/2014] [Indexed: 12/29/2022]
Abstract
Plasticity constitutes the basis of behavioral changes as a result of experience. It refers to neural network shaping and re-shaping at the global level and to synaptic contacts remodeling at the local level, either during learning or memory encoding, or as a result of acute or chronic pathological conditions. 'Plastic' brain reorganization after central nervous system lesions has a pivotal role in the recovery and rehabilitation of sensory and motor dysfunction, but can also be "maladaptive". Moreover, it is clear that brain reorganization is not a "static" phenomenon but rather a very dynamic process. Spinal cord injury immediately initiates a change in brain state and starts cortical reorganization. In the long term, the impact of injury - with or without accompanying therapy - on the brain is a complex balance between supraspinal reorganization and spinal recovery. The degree of cortical reorganization after spinal cord injury is highly variable, and can range from no reorganization (i.e. "silencing") to massive cortical remapping. This variability critically depends on the species, the age of the animal when the injury occurs, the time after the injury has occurred, and the behavioral activity and possible therapy regimes after the injury. We will briefly discuss these dependencies, trying to highlight their translational value. Overall, it is not only necessary to better understand how the brain can reorganize after injury with or without therapy, it is also necessary to clarify when and why brain reorganization can be either "good" or "bad" in terms of its clinical consequences. This information is critical in order to develop and optimize cost-effective therapies to maximize functional recovery while minimizing maladaptive states after spinal cord injury.
Collapse
Affiliation(s)
- K A Moxon
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA.
| | - A Oliviero
- Hospital Nacional de Parapléjicos, SESCAM, Finca la Peraleda s/n, 45071 Toledo, Spain
| | - J Aguilar
- Hospital Nacional de Parapléjicos, SESCAM, Finca la Peraleda s/n, 45071 Toledo, Spain
| | - G Foffani
- Hospital Nacional de Parapléjicos, SESCAM, Finca la Peraleda s/n, 45071 Toledo, Spain.
| |
Collapse
|
14
|
Bourke CH, Stowe ZN, Owens MJ. Prenatal antidepressant exposure: clinical and preclinical findings. Pharmacol Rev 2014; 66:435-65. [PMID: 24567054 DOI: 10.1124/pr.111.005207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pharmacological treatment of any maternal illness during pregnancy warrants consideration of the consequences of the illness and/or medication for both the mother and unborn child. In the case of major depressive disorder, which affects up to 10-20% of pregnant women, the deleterious effects of untreated depression on the offspring can be profound and long lasting. Progress has been made in our understanding of the mechanism(s) of action of antidepressants, fetal exposure to these medications, and serotonin's role in development. New technologies and careful study designs have enabled the accurate sampling of maternal serum, breast milk, umbilical cord serum, and infant serum psychotropic medication concentrations to characterize the magnitude of placental transfer and exposure through human breast milk. Despite this progress, the extant clinical literature is largely composed of case series, population-based patient registry data that are reliant on nonobjective means and retrospective recall to determine both medication and maternal depression exposure, and limited inclusion of suitable control groups for maternal depression. Conclusions drawn from such studies often fail to incorporate embryology/neurotransmitter ontogeny, appropriate gestational windows, or a critical discussion of statistically versus clinically significant. Similarly, preclinical studies have predominantly relied on dosing models, leading to exposures that may not be clinically relevant. The elucidation of a defined teratological effect or mechanism, if any, has yet to be conclusively demonstrated. The extant literature indicates that, in many cases, the benefits of antidepressant use during pregnancy for a depressed pregnant woman may outweigh potential risks.
Collapse
|
15
|
Money KM, Stanwood GD. Developmental origins of brain disorders: roles for dopamine. Front Cell Neurosci 2013; 7:260. [PMID: 24391541 PMCID: PMC3867667 DOI: 10.3389/fncel.2013.00260] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/01/2013] [Indexed: 01/11/2023] Open
Abstract
Neurotransmitters and neuromodulators, such as dopamine, participate in a wide range of behavioral and cognitive functions in the adult brain, including movement, cognition, and reward. Dopamine-mediated signaling plays a fundamental neurodevelopmental role in forebrain differentiation and circuit formation. These developmental effects, such as modulation of neuronal migration and dendritic growth, occur before synaptogenesis and demonstrate novel roles for dopaminergic signaling beyond neuromodulation at the synapse. Pharmacologic and genetic disruptions demonstrate that these effects are brain region- and receptor subtype-specific. For example, the striatum and frontal cortex exhibit abnormal neuronal structure and function following prenatal disruption of dopamine receptor signaling. Alterations in these processes are implicated in the pathophysiology of neuropsychiatric disorders, and emerging studies of neurodevelopmental disruptions may shed light on the pathophysiology of abnormal neuronal circuitry in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Kelli M Money
- Neuroscience Graduate Program, Vanderbilt University Nashville, TN, USA ; Vanderbilt Medical Scientist Training Program, Vanderbilt University Nashville, TN, USA
| | - Gregg D Stanwood
- Department of Pharmacology, Vanderbilt University Nashville, TN, USA ; Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University Nashville, TN, USA
| |
Collapse
|
16
|
Koizumi K, Nakajima H. Serotonin induces the migration of PC12 cells via the serotonin receptor 6/cAMP/ERK pathway. Biomed Rep 2013; 2:29-33. [PMID: 24649064 DOI: 10.3892/br.2013.203] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 10/25/2013] [Indexed: 02/02/2023] Open
Abstract
Serotonin (5-HT) functions as a chemoattractant that modulates neural migration during prenatal and early postnatal development. However, its molecular mechanism remains to be elucidated. The effect of 5-HT on neural cell migration was examined using PC12 neuron-like cell line. Transwell migration assay was used to determine the effect of 5-HT on PC12 cell migration. The results demonstrated that 5-HT and nerve growth factor (NGF) induced PC12 cell migration in a dose-dependent manner. Additionally, 5-HT receptor antagonists suggest that 5-HT-induced migration was mediated by serotonin receptor 6 (5-HT6), a Gs-protein coupled receptor that elevates the intercellular cAMP level. By contrast, antagonists of serotonin receptor 3 (5-HT3) did not show any effects on PC12 cell migration. Clozapine, an inhibitor of cAMP accumulation mediated by 5-HT6, significantly reduced the effect of 5-HT on the PC12 cell migration. An inhibitor of extracellular signal-regulated kinase (ERK) also suppressed migration. These results suggest that 5-HT induces PC12 cell migration by activating cAMP/ERK signaling pathways, which is mediated by 5-HT6 receptor.
Collapse
Affiliation(s)
- Keita Koizumi
- Center for Child Mental Development, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hideo Nakajima
- Department of Oncology, Ageo Central General Hospital, Ageo, Saitama, Japan ; Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
17
|
Georganta EM, Tsoutsi L, Gaitanou M, Georgoussi Z. δ-opioid receptor activation leads to neurite outgrowth and neuronal differentiation via a STAT5B-Gαi/o pathway. J Neurochem 2013; 127:329-41. [DOI: 10.1111/jnc.12386] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/27/2013] [Accepted: 07/29/2013] [Indexed: 12/25/2022]
Affiliation(s)
- Eirini-Maria Georganta
- Laboratory of Cellular Signalling and Molecular Pharmacology; Institute of Biosciences and Applications; National Centre for Scientific Research “Demokritos”; Athens Greece
| | - Lambrini Tsoutsi
- Laboratory of Cellular Signalling and Molecular Pharmacology; Institute of Biosciences and Applications; National Centre for Scientific Research “Demokritos”; Athens Greece
| | - Maria Gaitanou
- Laboratory of Cellular and Molecular Neurobiology; Hellenic Pasteur Institute; Athens Greece
| | - Zafiroula Georgoussi
- Laboratory of Cellular Signalling and Molecular Pharmacology; Institute of Biosciences and Applications; National Centre for Scientific Research “Demokritos”; Athens Greece
| |
Collapse
|
18
|
Anatomical and electrophysiological plasticity of locomotor networks following spinal transection in the salamander. Neurosci Bull 2013; 29:467-76. [PMID: 23893431 DOI: 10.1007/s12264-013-1363-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 05/19/2013] [Indexed: 01/09/2023] Open
Abstract
Recovery of locomotor behavior following spinal cord injury can occur spontaneously in some vertebrates, such as fish, urodele amphibians, and certain reptiles. This review provides an overview of the current status of our knowledge on the anatomical and electrophysiological changes occurring within the spinal cord that lead to, or are associated with the re-expression of locomotion in spinally-transected salamanders. A better understanding of these processes will help to devise strategies for restoring locomotor function in mammals, including humans.
Collapse
|
19
|
Vitalis T, Ansorge MS, Dayer AG. Serotonin homeostasis and serotonin receptors as actors of cortical construction: special attention to the 5-HT3A and 5-HT6 receptor subtypes. Front Cell Neurosci 2013; 7:93. [PMID: 23801939 PMCID: PMC3686152 DOI: 10.3389/fncel.2013.00093] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 05/27/2013] [Indexed: 12/15/2022] Open
Abstract
Cortical circuits control higher-order cognitive processes and their function is highly dependent on their structure that emerges during development. The construction of cortical circuits involves the coordinated interplay between different types of cellular processes such as proliferation, migration, and differentiation of neural and glial cell subtypes. Among the multiple factors that regulate the assembly of cortical circuits, 5-HT is an important developmental signal that impacts on a broad diversity of cellular processes. 5-HT is detected at the onset of embryonic telencephalic formation and a variety of serotonergic receptors are dynamically expressed in the embryonic developing cortex in a region and cell-type specific manner. Among these receptors, the ionotropic 5-HT3A receptor and the metabotropic 5-HT6 receptor have recently been identified as novel serotonergic targets regulating different aspects of cortical construction including neuronal migration and dendritic differentiation. In this review, we focus on the developmental impact of serotonergic systems on the construction of cortical circuits and discuss their potential role in programming risk for human psychiatric disorders.
Collapse
Affiliation(s)
- Tania Vitalis
- Laboratoire de Neurobiologie, ESPCI ParisTech, Centre National de la Recherche Scientifique-UMR 7637 Paris, France
| | | | | |
Collapse
|
20
|
Engel M, Smidt MP, van Hooft JA. The serotonin 5-HT3 receptor: a novel neurodevelopmental target. Front Cell Neurosci 2013; 7:76. [PMID: 23761731 PMCID: PMC3669892 DOI: 10.3389/fncel.2013.00076] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 05/06/2013] [Indexed: 01/28/2023] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT), next to being an important neurotransmitter, recently gained attention as a key-regulator of pre- and postnatal development in the mammalian central nervous system (CNS). Several receptors for 5-HT are expressed in the developing brain including a ligand-gated ion channel, the 5-HT3 receptor. Over the past years, evidence has been accumulating that 5-HT3 receptors are involved in the regulation of neurodevelopment by serotonin. Here, we review the spatial and temporal expression patterns of 5-HT3 receptors in the pre- and early postnatal rodent brain and its functional implications. First, 5-HT3 receptors are expressed on GABAergic interneurons in neocortex and limbic structures derived from the caudal ganglionic eminence. Mature inhibitory GABAergic interneurons fine-tune neuronal excitability and thus are crucial for the physiological function of the brain. Second, 5-HT3 receptors are expressed on specific glutamatergic neurons, Cajal-Retzius cells in the cortex and granule cells in the cerebellum, where they regulate morphology, positioning, and connectivity of the local microcircuitry. Taken together, the 5-HT3 receptor emerges as a potential key-regulator of network formation and function in the CNS, which could have a major impact on our understanding of neurodevelopmental disorders in which 5-HT plays a role.
Collapse
Affiliation(s)
- Mareen Engel
- Center for NeuroScience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
- Max Planck Institute of PsychiatryMunich, Germany
| | - Marten P. Smidt
- Center for NeuroScience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Johannes A. van Hooft
- Center for NeuroScience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| |
Collapse
|
21
|
van Kleef ESB, Gaspar P, Bonnin A. Insights into the complex influence of 5-HT signaling on thalamocortical axonal system development. Eur J Neurosci 2012; 35:1563-72. [PMID: 22607002 DOI: 10.1111/j.1460-9568.2012.8096.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The topographic organization of the thalamocortical axons (TCAs) in the barrel field (BF) in the rodent primary somatosensory cortex results from a succession of temporally and spatially precise developmental events. Prenatally, growth and guidance mechanisms enable TCAs to navigate through the forebrain and reach the cortex. Postnatally, TCAs grow into the cortex, and the refinement of their terminal arborization pattern in layer IV creates barrel-like structures. The combined results of studies performed over the past 20 years clearly show that serotonin (5-hydroxytryptamine; 5-HT) signaling modulates these pre- and early postnatal developmental processes. In this context, 5-HT signaling can purposely be described as 'modulating' rather than 'controlling' because developmental alterations of 5-HT synthesis, uptake or degradation either have a dramatic, moderate or no effect at all on TCA pathway and BF formation. In this review we summarize and compare the outcomes of diverse pharmacological and genetic manipulations of 5-HT signaling on TCA pathway and BF formation, in an attempt to understand these discrepancies.
Collapse
|
22
|
Abstract
In primary sensory neocortical areas of mammals, the distribution of sensory receptors is mapped with topographic precision and amplification in proportion to the peripheral receptor density. The visual, somatosensory and auditory cortical maps are established during a critical period in development. Throughout this window in time, the developing cortical maps are vulnerable to deleterious effects of sense organ damage or sensory deprivation. The rodent barrel cortex offers an invaluable model system with which to investigate the mechanisms underlying the formation of topographic maps and their plasticity during development. Five rows of mystacial vibrissa (whisker) follicles on the snout and an array of sinus hairs are represented by layer IV neural modules ('barrels') and thalamocortical axon terminals in the primary somatosensory cortex. Perinatal damage to the whiskers or the sensory nerve innervating them irreversibly alters the structural organization of the barrels. Earlier studies emphasized the role of the sensory periphery in dictating whisker-specific brain maps and patterns. Recent advances in molecular genetics and analyses of genetically altered mice allow new insights into neural pattern formation in the neocortex and the mechanisms underlying critical period plasticity. Here, we review the development and patterning of the barrel cortex and the critical period plasticity.
Collapse
Affiliation(s)
- Reha S Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201-1075, USA.
| | | |
Collapse
|
23
|
Neonatal fluoxetine exposure alters motor performances of adolescent rats. Dev Neurobiol 2012; 72:1122-32. [DOI: 10.1002/dneu.20942] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/16/2011] [Accepted: 06/17/2011] [Indexed: 11/07/2022]
|
24
|
Trakhtenberg EF, Goldberg JL. The role of serotonin in axon and dendrite growth. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012; 106:105-26. [PMID: 23211461 DOI: 10.1016/b978-0-12-407178-0.00005-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The neurotransmitter serotonin (5-hydroxytryptamine, 5-HT) plays multiple roles in the enteric, peripheral, and central nervous systems (CNS). Although its most prominent biological function is as a signal transmission messenger from pre- to postsynaptic neurons, other roles such as shaping brain development and regulating neurite growth have also been described. Here, we review the less well-studied role of 5-HT as a modulator of neurite growth. 5-HT has been shown to regulate neurite growth in multiple systems and species, including in the mammalian CNS. 5-HT predominantly appears to suppress neurite growth, but depending on the model system and 5-HT receptor subtype, in rare cases, it may promote neurite outgrowth and elongation. Failure of axon regeneration in the adult mammalian CNS is a major problem in multiple diseases, and understanding how 5-HT receptors signal opposing effects on neurite growth may lead to novel neuroregenerative therapies, by targeting either 5-HT receptors or their downstream signaling pathways.
Collapse
Affiliation(s)
- Ephraim F Trakhtenberg
- Bascom Palmer Eye Institute, Interdisciplinary Stem Cell Institute, Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | | |
Collapse
|
25
|
Liao CC, Lee LJ. Neonatal fluoxetine exposure affects the action potential properties and dendritic development in cortical subplate neurons of rats. Toxicol Lett 2011; 207:314-21. [PMID: 21986067 DOI: 10.1016/j.toxlet.2011.09.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 09/26/2011] [Accepted: 09/26/2011] [Indexed: 01/26/2023]
Abstract
Selective serotonin reuptake inhibitor (SSRI)-type antidepressants might be given to depressive pregnant women and the developing fetuses are thus exposed to these drugs. Since serotonin plays important roles in the maturation of the nervous system, early SSRI exposure might influence the fetal brain development. To test this hypothesis, we treated the neonatal rat pups with fluoxetine (Flx) from the day of birth to postnatal day (P) 4, comparable to the third trimester of human gestation, and observed the physiological and morphological features of subplate neurons (SPns), a group of cells important for early cortical development and vulnerable to neonatal neural insults. Using whole-cell patch-clamp recording technique, we examined the passive membrane properties and characteristics of action potential (AP). In SPns of Flx-treated rats, the rheobase for generating an AP was increased and the width of APs was reduced, especially in the falling phase. In the morphological aspect, the dendritic remodeling of SPns including dendritic branching, elongation and pruning were affected by early Flx treatment. Together, our results demonstrate that the teratogenic effect of early SSRI exposure on the structure and function of developing SPns and these changes may lead to undesired brain activity and distorted behaviors later in life.
Collapse
Affiliation(s)
- Chun-Chieh Liao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan
| | | |
Collapse
|
26
|
Ferreira TA, Iacono LL, Gross CT. Serotonin receptor 1A modulates actin dynamics and restricts dendritic growth in hippocampal neurons. Eur J Neurosci 2010; 32:18-26. [PMID: 20561047 DOI: 10.1111/j.1460-9568.2010.07283.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mice lacking serotonin receptor 1A (Htr1a) display increased anxiety behavior that depends on the expression of the receptor in the forebrain during the third to fifth postnatal weeks. Within the forebrain, Htr1a is prominently expressed in the soma and dendrites of CA1 pyramidal neurons of the hippocampus and these cells undergo rapid dendritic growth and synapse formation during this period. Consistent with a possible role of Htr1a in synaptic maturation, CA1 pyramidal neurons in the knockout mice show increased ramification of oblique dendrites. These findings suggest that Htr1a may shape hippocampal circuits by directly modulating dendritic growth. Here we show that pharmacological blockade of the receptor during the third to fifth postnatal weeks is sufficient to reproduce the increased branching of oblique dendrites seen in knockout mice. Using dissociated hippocampal cultures we demonstrate that serotonin functions through Htr1a to attenuate the motility of dendritic growth cones, reduce their content of filamentous actin and alter their morphology. These findings suggest that serotonin modulates actin cytoskeletal dynamics in hippocampal neurons during a limited developmental period to restrict dendritic growth and achieve a long-term adjustment of neural connectivity.
Collapse
|
27
|
Roles of serotonin 5-HT3 receptor in the formation of dendrites and axons in the rat cerebral cortex: An in vitro study. Neurosci Res 2010; 66:22-9. [DOI: 10.1016/j.neures.2009.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 09/09/2009] [Accepted: 09/14/2009] [Indexed: 11/17/2022]
|
28
|
Bortolato M, Chen K, Shih JC. The Degradation of Serotonin: Role of MAO. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2010. [DOI: 10.1016/s1569-7339(10)70079-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
29
|
The role of glial cells in influencing neurite extension by dorsal root ganglion cells. ACTA ACUST UNITED AC 2009; 6:19-29. [PMID: 20025817 DOI: 10.1017/s1740925x09990433] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
When pretreated with pertussis toxin (PTX), the neurites of adult rat dorsal root ganglion (DRG) cells in mixed cell cultures retract over a period of 2 h following the initial stimulus of removal from the cell culture incubator for brief periods of observation. The purpose of this investigation was to determine whether this PTX-dependent response was specific to any one of the three subpopulations of DRG neurons. However, no neurite retraction response was observed in neuron-enriched populations of cells, or in cultures enriched in isolectin B4 (IB4)-positive neurons or in IB4-negative neurons. But, the addition of non-neuronal cells, and/or medium conditioned by non-neuronal cells, was sufficient to restore the PTX-dependent neurite retraction response, but only in large diameter IB4-negative neurons. In conclusion, we have identified a regulatory response, mediated by Gi/o-proteins, which prevents retraction of neurites in large diameter IB4-negative cells of adult rat DRG. The non-neuronal cells of adult rat DRG constitutively release factor/s that can stimulate neurite retraction of a subset of isolated DRG neurons, but this property of non-neuronal cells is only observed when the Gi/o-proteins of large diameter IB4-negative cells are inhibited.
Collapse
|
30
|
Abstract
Signaling from G(i/o)-coupled G protein-coupled receptors (GPCRs), such as the serotonin 1B, cannabinoid 1, and dopamine D2 receptors, inhibits cAMP production by adenylyl cyclases and activates protein kinases, such as Src, mitogen-activated protein kinases 1 and 2, and Akt. Activation of these protein kinases results in stimulation of neurite outgrowth in the central nervous system (CNS) and in neuronal cell lines. This Connections Map traces downstream signaling pathways from G(i/o)-coupled GPCRs to key protein kinases and key transcription factors involved in neuronal differentiation. Components in the Science Signaling Connections Map are linked to Nature Molecule Pages. This interoperability provides ready access to detail that includes information about specific states for the nodes.
Collapse
Affiliation(s)
- Avi Ma'ayan
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | |
Collapse
|
31
|
Masuho I, Mototani Y, Sahara Y, Asami J, Nakamura S, Kozasa T, Inoue T. Dynamic expression patterns of G protein-regulated inducer of neurite outgrowth 1 (GRIN1) and its colocalization with Galphao implicate significant roles of Galphao-GRIN1 signaling in nervous system. Dev Dyn 2009; 237:2415-29. [PMID: 18729205 DOI: 10.1002/dvdy.21686] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
GRIN1 (Gprin 1) is a signaling molecule coexpression of which with constitutively active form of Galphao can stimulate neurite extensions in Neuro2a cells, yet its in vivo roles remain elusive. Here, we examine expression profiles of GRIN1 during mouse development by in situ hybridization (ISH) and immunohistochemistry. ISH analysis revealed that GRIN1 expression was limited to the nervous system at all developmental stages tested: in the central nervous system, GRIN1 expression occurred within the entire embryonic mantle zones, while it became restricted to sets of nuclei at postnatal to adult stages. Immunohistochemistry using a GRIN1-specific antibody demonstrated that GRIN1 colocalized with Galphao at neuronal dendrites and axons, but it was not detected in glial cells. These results suggest that Galphao-GRIN1 pathway could mediate significant roles in neuronal migration and differentiation at embryonic stages and exert functions in wiring and/or maintenance of specific neural circuitries at postnatal to adult stages.
Collapse
Affiliation(s)
- Ikuo Masuho
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Illinois 60612, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Roychowdhury S, Rasenick MM. Submembraneous microtubule cytoskeleton: regulation of microtubule assembly by heterotrimeric Gproteins. FEBS J 2008; 275:4654-63. [PMID: 18754776 PMCID: PMC2782913 DOI: 10.1111/j.1742-4658.2008.06614.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Heterotrimeric Gproteins participate in signal transduction by transferring signals from cell surface receptors to intracellular effector molecules. Gproteins also interact with microtubules and participate in microtubule-dependent centrosome/chromosome movement during cell division, as well as neuronal differentiation. In recent years, significant progress has been made in our understanding of the biochemical/functional interactions between Gprotein subunits (alpha and betagamma) and microtubules, and the molecular details emerging from these studies suggest that alpha and betagamma subunits of Gproteins interact with tubulin/microtubules to regulate the assembly/dynamics of microtubules, providing a novel mechanism for hormone- or neurotransmitter-induced rapid remodeling of cytoskeleton, regulation of the mitotic spindle for centrosome/chromosome movements in cell division, and neuronal differentiation in which structural plasticity mediated by microtubules is important for appropriate synaptic connections and signal transmission.
Collapse
Affiliation(s)
- Sukla Roychowdhury
- Department of Biological Sciences, University of Texas, El Paso, TX, USA.
| | | |
Collapse
|
33
|
Bras H, Gaytán SP, Portalier P, Zanella S, Pásaro R, Coulon P, Hilaire G. Prenatal activation of 5-HT2A receptor induces expression of 5-HT1B receptor in phrenic motoneurons and alters the organization of their premotor network in newborn mice. Eur J Neurosci 2008; 28:1097-107. [PMID: 18783379 DOI: 10.1111/j.1460-9568.2008.06407.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In newborn mice of the control [C3H/HeJ (C3H)] and monoamine oxidase A-deficient (Tg8) strains, in which levels of endogenous serotonin (5-HT) were drastically increased, we investigated how 5-HT system dysregulation affected the maturation of phrenic motoneurons (PhMns), which innervate the diaphragm. First, using immunocytochemistry and confocal microscopy, we observed a 5-HT(2A) receptor (5-HT(2A)-R) expression in PhMns of both C3H and Tg8 neonates at the somatic and dendritic levels, whereas 5-HT(1B) receptor (5-HT(1B)-R) expression was observed only in Tg8 PhMns at the somatic level. We investigated the interactions between 5-HT(2A)-R and 5-HT(1B)-R during maturation by treating pregnant C3H mice with a 5-HT(2A)-R agonist (2,5-dimethoxy-4-iodoamphetamine hydrochloride). This pharmacological overactivation of 5-HT(2A)-R induced a somatic expression of 5-HT(1B)-R in PhMns of their progeny. Conversely, treatment of pregnant Tg8 mice with a 5-HT(2A)-R antagonist (ketanserin) decreased the 5-HT(1B)-R density in PhMns of their progeny. Second, using retrograde transneuronal tracing with rabies virus injected into the diaphragm of Tg8 and C3H neonates, we studied the organization of the premotor network driving PhMns. The interneuronal network monosynaptically connected to PhMns was much more extensive in Tg8 than in C3H neonates. However, treatment of pregnant C3H mice with 2,5-dimethoxy-4-iodoamphetamine hydrochloride switched the premotoneuronal network of their progeny from a C3H- to a Tg8-like pattern. These results show that a prenatal 5-HT excess affects, via the overactivation of 5-HT(2A)-R, the expression of 5-HT(1B)-R in PhMns and the organization of their premotor network.
Collapse
Affiliation(s)
- Hélène Bras
- Laboratoire Plasticité et Physio-Pathologie de la Motricité, UMR 6196 CNRS, Université de la Méditerranée, Marseille, France.
| | | | | | | | | | | | | |
Collapse
|
34
|
Kichula EA, Huntley GW. Developmental and comparative aspects of posterior medial thalamocortical innervation of the barrel cortex in mice and rats. J Comp Neurol 2008; 509:239-58. [PMID: 18496871 DOI: 10.1002/cne.21690] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The thalamocortical projection to the rodent barrel cortex consists of inputs from the ventral posterior medial (VPM) and posterior medial (POm) nuclei that terminate in largely nonoverlapping territories in and outside of layer IV. This projection in both rats and mice has been used extensively to study development and plasticity of highly organized synaptic circuits. Whereas the VPM pathway has been well characterized in both rats and mice, organization of the POm pathway has only been described in rats, and no studies have focused exclusively on the development of the POm projection. Here, using transport of Phaseolus vulgaris leucoagglutinin(PHA-L) or carbocyanine dyes, we characterize the POm thalamocortical innervation of adult mouse barrel cortex and describe its early postnatal development in both mice and rats. In adult mice, POm inputs form a dense plexus in layer Va that extends uniformly underneath layer IV barrels and septa. Innervation of layer IV is very sparse; a clear septal innervation pattern is evident only at the layer IV/Va border. This pattern differs subtly from that described previously in rats. Developmentally, in both species, POm axons are present in barrel cortex at birth. In mice, they occupy layer IV as it differentiates, whereas in rats, POm axons do not enter layer IV until 1-2 days after its emergence from the cortical plate. In both species, arbors undergo progressive and directed growth. However, no layer IV septal innervation pattern emerges until several days after the cytoarchitectonic appearance of barrels and well after the emergence of whisker-related clusters of VPM thalamocortical axons. The mature pattern resolves earlier in rats than in mice. Taken together, these data reveal anatomical differences between mice and rats in the development and organization of POm inputs to barrel cortex, with implications for species differences in the nature and plasticity of lemniscal and paralemniscal information processing.
Collapse
Affiliation(s)
- Elizabeth A Kichula
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, New York 10029-6574, USA
| | | |
Collapse
|
35
|
Glutamate transporters regulate lesion-induced plasticity in the developing somatosensory cortex. J Neurosci 2008; 28:4995-5006. [PMID: 18463253 DOI: 10.1523/jneurosci.0861-08.2008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glutamate transporters are involved in neural differentiation, neuronal survival, and synaptic transmission. In the present study, we examined glutamate transporter 1 (GLT1) expression in the neonatal somatosensory cortex of C57BL/6 mice, and pursued its role in somatosensory development by comparing barrel development between GLT1 knock-out and control mice. During the first few neonatal days, a critical period for barrels, GLT1 expression is strikingly upregulated in cortical astrocytes, whereas it was downregulated in neuronal elements to below the detection threshold. GLT1 knock-out neonates developed normally in terms of body growth, cortical histoarchitecture, barrel formation, and critical period termination. However, when row C whiskers were lesioned during the critical period, reduction of lesioned row C barrels and reciprocal expansion of intact row B/D barrels were both milder in GLT1 knock-out mice than in control littermates. Accordingly, the map plasticity index, calculated as (B + D)/2C, was significantly lowered in GLT1 knock-out mice. We also found that extracellular glutamate levels in the neonatal somatosensory cortex were significantly elevated in GLT1 knock-out mice. Diminished lesion-induced plasticity was further found in mutant mice lacking glutamate-aspartate transporter (GLAST), an astrocyte-specific glutamate transporter throughout development. Therefore, glutamate transporters regulate critical period plasticity by enhancing expansion of active barrels and shrinkage of inactive barrels. Because cortical contents of glutamate receptors and GLAST were unaltered in GLT1 knock-out mice, this action appears to be mediated, at least partly, by keeping the ambient glutamate level low. Considering an essential role of glutamate receptors in the formation of whisker-related thalamocortical synapse patterning, glutamate transporters thus facilitate their activity-dependent remodeling.
Collapse
|
36
|
Bromberg KD, Iyengar R, He JC. Regulation of neurite outgrowth by G(i/o) signaling pathways. FRONT BIOSCI-LANDMRK 2008; 13:4544-57. [PMID: 18508528 DOI: 10.2741/3022] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neurogenesis is a long and winding journey. A neural progenitor cell migrates long distances, differentiates by forming a single axon and multiple dendrites, undergoes maturation, and ultimately survives. The initial formation of neurites during neuronal differentiation, commonly referred to as "neurite outgrowth," can be induced by a large repertoire of signals that stimulate an array of receptors and downstream signaling pathways. The G(i/o) family of heterotrimeric G-proteins are abundantly expressed in the brain and enriched at neuronal growth cones. Recent evidence has uncovered several G(i/o)-coupled receptors that induce neurite outgrowth and has begun to elucidate the underlying molecular mechanisms. Emerging data suggests that signals from several G(i/o)-coupled receptors converge at the transcription factor STAT3 to regulate neurite outgrowth and at Rac1 and Cdc42 to regulate cytoskeletal reorganization. Physiologically, signaling through G(i/o)-coupled cannabinoid receptors is critical for pro percentral nervous system development. As the mechanisms by which G(i/o)-coupled receptors regulate neurite outgrowth are clarified, it is becoming evident that modulating signals from G(i/o) and their receptors has great potential for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Kenneth D Bromberg
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | |
Collapse
|
37
|
|
38
|
Bonnin A, Torii M, Wang L, Rakic P, Levitt P. Serotonin modulates the response of embryonic thalamocortical axons to netrin-1. Nat Neurosci 2007; 10:588-97. [PMID: 17450135 DOI: 10.1038/nn1896] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Accepted: 03/19/2007] [Indexed: 11/09/2022]
Abstract
Modifying serotonin (5-HT) abundance in the embryonic mouse brain disrupts the precision of sensory maps formed by thalamocortical axons (TCAs), suggesting that 5-HT influences their growth. We investigated the mechanism by which 5-HT influences TCAs during development. 5-HT(1B) and 5-HT(1D) receptor expression in the fetal forebrain overlaps with that of the axon guidance receptors DCC and Unc5c. In coculture assays, axons originating from anterior and posterior halves of the embryonic day 14.5 dorsal thalamus responded differently to netrin-1, reflecting the patterns of DCC and Unc5c expression. 5-HT converts the attraction exerted by netrin-1 on posterior TCAs to repulsion. Pharmacological manipulation of 5-HT(1B/1D) receptors and intracellular cAMP showed the signaling cascade through which this modulation occurs. An in vivo correlate of altered TCA pathfinding was obtained by transient manipulation of 5-HT(1B/1D) receptor expression abundance in the dorsal thalamus by in utero electroporation. These data demonstrate that serotonergic signaling has a previously unrecognized role in the modulation of axonal responsiveness to a classic guidance cue.
Collapse
Affiliation(s)
- Alexandre Bonnin
- Department of Pharmacology, Vanderbilt University, 465 21st Ave. South, Nashville, Tennessee 37232, USA
| | | | | | | | | |
Collapse
|
39
|
He JC, Neves SR, Jordan JD, Iyengar R. Role of the Go/i signaling network in the regulation of neurite outgrowth. Can J Physiol Pharmacol 2007; 84:687-94. [PMID: 16998532 DOI: 10.1139/y06-025] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Neurite outgrowth is a complex differentiation process stimulated by many neuronal growth factors and transmitters and by electrical activity. Among these stimuli are ligands for G-protein-coupled receptors (GPCR) that function as neurotransmitters. The pathways involved in GPCR-triggered neurite outgrowth are not fully understood. Many of these receptors couple to Galphao, one of the most abundant proteins in the neuronal growth cones. We have studied the Go signaling network involved in neurite outgrowth in Neuro2A cells. Galphao can induce neurite outgrowth. The CB1 cannabinoid receptor, a Go/i-coupled receptor expressed endogenously in Neuro2A cells, triggers neurite outgrowth by activating Rap1, which promotes the Galphao-stimulated proteasomal degradation of Rap1GAPII. CB1-receptor-mediated Rap1 activation leads to the activation of a signaling network that includes the small guanosine triphosphate (GTP)ases Ral and Rac, the protein kinases Src, and c-Jun N-terminal kinase (JNK), which converge onto the activation of signal transducer and activator of transcription 3 (Stat3), a key transcription factor that mediates the gene expression process of neurite outgrowth in Neuro2A cells. This review describes current findings from our laboratory and also discusses alternative pathways that Go/i might mediate to trigger neurite outgrowth. We also analyze the role neurotransmitters, which stimulate Go/i to activate a complex signaling network controlling neurite outgrowth, play in regeneration after neuronal injury.
Collapse
Affiliation(s)
- John Cijiang He
- Department of Pharmacology and Biological Chemistry, Mount Sinai School of Medicine, One Gustave L levy Place, New York, NY 10029, USA.
| | | | | | | |
Collapse
|
40
|
Boylan CB, Blue ME, Hohmann CF. Modeling early cortical serotonergic deficits in autism. Behav Brain Res 2006; 176:94-108. [PMID: 17034875 PMCID: PMC2570481 DOI: 10.1016/j.bbr.2006.08.026] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Revised: 08/15/2006] [Accepted: 08/17/2006] [Indexed: 10/24/2022]
Abstract
Autism is a developmental brain disorder characterized by deficits in social interaction, language and behavior. Brain imaging studies demonstrate increased cerebral cortical volumes and micro- and macro-scopic neuroanatomic changes in children with this disorder. Alterations in forebrain serotonergic function may underlie the neuroanatomic and behavioral features of autism. Serotonin is involved in neuronal growth and plasticity and these actions are likely mediated via serotonergic and glutamatergic receptors. Few animal models of autism have been described that replicate both etiology and pathophysiology. We report here on a selective serotonin (5-HT) depletion model of this disorder in neonatal mice that mimics neurochemical and structural changes in cortex and, in addition, displays a behavioral phenotype consistent with autism. Newborn male and female mice were depleted of forebrain 5-HT with injections of the serotonergic neurotoxin, 5,7-dihydroxytryptamine (5,7-DHT), into the bilateral medial forebrain bundle (mfb). Behavioral testing of these animals as adults revealed alterations in social, sensory and stereotypic behaviors. Lesioned mice showed significantly increased cortical width. Serotonin immunocytochemistry showed a dramatic long-lasting depletion of 5-HT containing fibers in cerebral cortex until postnatal day (PND) 60. Autoradiographic binding to high affinity 5-HT transporters was significantly but transiently reduced in cerebral cortex of 5,7-DHT-depleted mice. AMPA glutamate receptor binding was decreased at PND 15. We hypothesize that increased cerebral cortical volume and sensorimotor, cognitive and social deficits observed in both 5-HT-depleted animals and in individuals with autism, may be the result of deficiencies in timely axonal pruning to key cerebral cortical areas.
Collapse
Affiliation(s)
- Carolyn B. Boylan
- Division of Neonatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Mary E. Blue
- Kennedy Krieger Research Institute and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- *Corresponding Author: Mary E. Blue, PhD., Kennedy Krieger Research Institute, 707 N. Broadway, Baltimore, MD 21205. Telephone 443-923-2693, Fax 443-923-2695,
| | | |
Collapse
|
41
|
Homma K, Kitamura Y, Ogawa H, Oka K. Serotonin induces the increase in intracellular Ca2+ that enhances neurite outgrowth in PC12 cells via activation of 5-HT3 receptors and voltage-gated calcium channels. J Neurosci Res 2006; 84:316-25. [PMID: 16688720 DOI: 10.1002/jnr.20894] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
As a neurotransmitter and neuromodulator, serotonin (5-HT) influences neuronal outgrowth in the nervous systems of several species. In PC12 cells, 5-HT is known to have neuritogenic effects, although the signal transduction pathway responsible for these effects is not understood. In this study, we hypothesized that a 5-HT-induced increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) could be involved in mediating the effects of 5-HT. Application of 5-HT to PC12 cells enhanced nerve growth factor (NGF)-induced neurite outgrowth in a dose-dependent manner, and the sensitivity of this neuritogenic effect was increased in differentiated PC12 cells. In accordance, an increase in [Ca(2+)](i) was observed following application of 5-HT in differentiated PC12 cells. This increase was amplified by further NGF treatment. 5-HT-induced increases in [Ca(2+)](i) were inhibited by MDL 72222, a selective 5-HT(3) receptor antagonist, and nifedipine, an L-type calcium channel blocker, but not by ketanserin, a 5-HT(2) receptor antagonist, or thapsigargin, a specific inhibitor of endoplasmic reticulum Ca(2+)-ATPase. These pharmacological tests indicated that 5-HT-induced increases in [Ca(2+)](i) are mediated by activation of voltage-gated calcium channels via 5-HT(3) receptors and that 5-HT-induced increases in [Ca(2+)](i) are likely to be independent of activation of 5-HT(2) receptors in PC12 cells. Furthermore, the neuritogenic effect of 5-HT was suppressed by MDL 72222, nifedipine, calmodulin (CaM) inhibitor, and calcineurin inhibitors. Taken together, our results indicate that 5-HT-induced increases in [Ca(2+)](i), which are mediated via 5-HT(3) receptors and L-type calcium channels in PC12 cells, and subsequent activation of CaM and calcineurin enhance NGF-induced neurite outgrowth.
Collapse
Affiliation(s)
- Kohei Homma
- Center for Life Science and Technology, School of Fundamental Science and Technology, Graduate School of Science and Technology, Keio University, Yokohama, Kanagawa, Japan
| | | | | | | |
Collapse
|
42
|
Altamura C, Dell'Acqua ML, Moessner R, Murphy DL, Lesch KP, Persico AM. Altered Neocortical Cell Density and Layer Thickness in Serotonin Transporter Knockout Mice: A Quantitation Study. Cereb Cortex 2006; 17:1394-401. [PMID: 16905592 DOI: 10.1093/cercor/bhl051] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The neurotransmitter serotonin (5-HT) plays morphogenetic roles during development, and their alteration could contribute to autism pathogenesis in humans. To further characterize 5-HT's contributions to neocortical development, we assessed the thickness and neuronal cell density of various cerebral cortical areas in serotonin transporter (5-HTT) knockout (ko) mice, characterized by elevated extracellular 5-HT levels. The thickness of layer IV is decreased in 5-HTT ko mice compared with wild-type (wt) mice. The overall effect on cortical thickness, however, depends on the genetic background of the mice. Overall cortical thickness is decreased in many cortical areas of 5-HTT ko mice with a mixed c129-CD1-C57BL/6J background. Instead, 5-HTT ko mice backcrossed into the C57BL/6J background display increases in supragranular and infragranular layers, which compensate entirely for decreased layer IV thickness, resulting in unchanged or even enhanced cortical thickness. Moreover, significant increases in neuronal cell density are found in 5-HTT ko mice with a C57BL/6J background (wt:hz:ko ratio = 1.00:1.04:1.17) but not in the mixed c129-CD1-C57BL/6J 5-HTT ko animals. These results provide evidence of 5-HTT gene effects on neocortical morphology in epistatic interaction with genetic variants at other loci and may model the effect of functional 5-HTT gene variants on neocortical development in autism.
Collapse
Affiliation(s)
- C Altamura
- Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Via Longoni 83, I-00155 Rome, Italy
| | | | | | | | | | | |
Collapse
|
43
|
Persico AM, Di Pino G, Levitt P. Multiple receptors mediate the trophic effects of serotonin on ventroposterior thalamic neurons in vitro. Brain Res 2006; 1095:17-25. [PMID: 16701576 DOI: 10.1016/j.brainres.2006.04.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Revised: 03/23/2006] [Accepted: 04/03/2006] [Indexed: 11/30/2022]
Abstract
Serotonin (5-HT) exerts prominent morphogenetic roles during development. For example, somatosensory cortical barrel formation is altered in mouse models characterized by excessive extracellular 5-HT, suggesting that 5-HT affects development of thalamic afferents and/or neocortical target regions. The present study assessed 5-HT effects in primary cultures of fetal ventroposterior thalamic (VPT) neurons. 5-HT produces concentration-dependent trophic effects, with impressive 59% and 106% peak increases in total neurite length and number of branching points, respectively, at a dose of 30 microM 5-HT. The exposure of VPT neurons to specific 5-HT receptor agonists 8-OH-DPAT (5-HT(1A)), CGS-12066A (5-HT(1B)), DOI (5-HT(2A/2C)), and m-CPBG (5-HT(3)), enhances primary neurite length and number of branching points with rank-order potency 5-HT(1B) > 5-HT(2A/2C) = 5-HT(3) > 5-HT(1A) = vehicle. Trophic 5-HT effects on embryonic VPT neurons are thus much more prominent than previously reported, and can be mediated by multiple 5-HT receptor subtypes.
Collapse
Affiliation(s)
- Antonio M Persico
- Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Via Emilio Longoni 83, I-00155 Rome, Italy.
| | | | | |
Collapse
|
44
|
Zhou FC, Sari Y, Powrozek TA. Fetal Alcohol Exposure Reduces Serotonin Innervation and Compromises Development of the Forebrain Along the Serotonergic Pathway. Alcohol Clin Exp Res 2006; 29:141-9. [PMID: 15654302 DOI: 10.1097/01.alc.0000150636.19677.6f] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND We reported previously that a moderate level of fetal alcohol treatment reduces the birth, maturation, and migration of serotonin (5-HT) neurons at embryonic days 11 to 15 (E11-E15). Because 5-HT is known as a differentiation signal for forebrain development, we investigated whether alcohol affects 5-HT innervation to the developing brain and how the target brain areas grow as they receive 5-HT innervation between E15 and E18. METHODS Pregnant dams were divided into three groups and treated from E7 to E15 or E18 with one of the following conditions: (1) liquid diet that contained 25% ethanol-derived calories (ALC), (2) isocaloric liquid diet pair-fed (PF), or (3) chow fed (Chow). The 5-HT immunostained (5-HT-IM) fibers and size of brain areas were examined as an index of growth along the ascending 5-HT pathway. RESULT We found that 5-HT-IM fibers innervate the brain regions specifically under active differentiation and that there were three sets of correlated dysmorphology in the ALC group as compared with those of the PF and Chow groups. The three sets are as follows: (1) fewer 5-HT-IM fibers in the medial forebrain bundle and along the projecting pathway through the hypothalamus, septal nucleus, frontal and parietal cortices, and subiculum/hippocampus; (2) underdevelopment of the brain regions along 5-HT fiber projections; and (3) underdevelopment of somatosensory thalamocortical projections, which are known to transiently express 5-HT transporters and to be regulated by 5-HT. No such differences were found between the PF and Chow groups. CONCLUSION We found that fewer 5-HT fibers grew in the embryos that were exposed to alcohol. As forebrain regions differentiated along the 5-HT projection, we found two reductions: (1) the growth of brain regions along 5-HT projection and (2) the growth of the thalamocortical sensory projection regulated by 5-HT. The reduced 5-HT innervation is in agreement with our previous observation of fewer 5-HT neurons. The subsequent retardation of forebrain growth and sensory thalamocortical fibers along the pathway of reduced 5-HT projection is consistent with the role of 5-HT as a signal for forebrain differentiation.
Collapse
Affiliation(s)
- Feng C Zhou
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | |
Collapse
|
45
|
Bonnin A, Peng W, Hewlett W, Levitt P. Expression mapping of 5-HT1 serotonin receptor subtypes during fetal and early postnatal mouse forebrain development. Neuroscience 2006; 141:781-794. [PMID: 16824687 DOI: 10.1016/j.neuroscience.2006.04.036] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Revised: 04/03/2006] [Accepted: 04/05/2006] [Indexed: 11/20/2022]
Abstract
Serotonin (5-HT) is implicated in several aspects of brain development, yet the ontogenetic expression patterns of 5-HT receptors responsible for transducing specific effects have largely not been characterized. Fifteen different 5-HT receptor genes have been cloned; therefore any spatial and/or temporal combination of their developmental expression could mediate a wide array of 5-HT effects. We undertook a detailed analysis of expression mapping of the Gi/o-coupled 5-HT1 (5-HT1A, 1B, 1D and 1F) receptor subtypes in the fetal and early postnatal mouse forebrain. Using receptor subtype-specific riboprobes and in situ hybridization, we observed that all 5-HT1 receptor subtypes are expressed as early as embryonic day (E) 14.5 in the forebrain, typically in gradients within specific structures. Among 5-HT1 receptors, the 5-HT1A receptor transcript is expressed densely in E14.5-16.5 thalamus, in hippocampus, and in a medial to lateral gradient in cortex, whereas the 5-HT1B receptor mRNA is expressed in more lateral parts of the dorsal thalamus and in the striatum at these ages. The 5-HT1D receptor transcript, which also is expressed heavily in E14.5-E16.5 thalamus, appears to be down-regulated at birth. The 5-HT1F receptor transcript is present in proliferative regions such as the cortical ventricular zone, ganglionic eminences, and medial aspects of the thalamus at E14.5-16.5, and otherwise presents similarities to the expression patterns of 5-HT1B and 1D receptor transcripts. Overall, the 5-HT1 subfamily of Gi/o-coupled 5-HT receptors displays specific and dynamic expression patterns during embryonic forebrain development. Moreover, all members of the 5-HT1 receptor class are strongly and transiently expressed in the embryonic dorsal thalamus, which suggests a potential role for serotonin in early thalamic development.
Collapse
Affiliation(s)
- A Bonnin
- Department of Pharmacology, Vanderbilt University, 8114 MRBIII, 465 21st Avenue South, Nashville, TN 37232-8548, USA; Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Box 40 Peabody, 230 Appleton Place, Nashville, TN 37203, USA
| | - W Peng
- Department of Psychiatry, Vanderbilt School of Medicine, 1601 23rd Avenue South, Nashville, TN 37232-8645, USA
| | - W Hewlett
- Department of Psychiatry, Vanderbilt School of Medicine, 1601 23rd Avenue South, Nashville, TN 37232-8645, USA; Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Box 40 Peabody, 230 Appleton Place, Nashville, TN 37203, USA
| | - P Levitt
- Department of Pharmacology, Vanderbilt University, 8114 MRBIII, 465 21st Avenue South, Nashville, TN 37232-8548, USA; Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Box 40 Peabody, 230 Appleton Place, Nashville, TN 37203, USA.
| |
Collapse
|
46
|
Lane RD, Chiaia NL, Kesterson KL, Rhoades RW, Mooney RD. Boundary-limited serotonergic influences on pattern organization in rat sensory cortex. Neurosci Lett 2005; 395:165-9. [PMID: 16325338 DOI: 10.1016/j.neulet.2005.10.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Revised: 10/15/2005] [Accepted: 10/26/2005] [Indexed: 11/19/2022]
Abstract
In neonatal rodents, elevated levels of cortical serotonin (5-HT) blur the normally segmented vibrissae-related pattern of thalamocortical afferents (TCAs) in the posteromedial barrel subfield (PMBSF) of primary somatosensory cortex. We employed 5-HT immunocytochemistry or anterograde transport of 1'1'-dioctadecyl-3,3,3',3' tetramethyl-indocarbocyanin (Di-I) to label TCA arbors to study the effects of 5-HT manipulations on space occupied by TCAs within the PMBSF and the total area labeled. In rats treated to increase cortical 5-HT from birth to postnatal day (P) 6, the percentage of PMBSF area occupied by terminal labeling was significantly higher from that in controls (79.0% versus 23.7%, P < 0.05) for the highest levels of cortical 5-HT and was raised, although not significantly, for lower levels of 5-HT. The TCA coverage was significantly correlated with treatment dose. In animals exposed to a selective 5-HT1B agonist, 5-nonyloxytryptamine, or elevated endogenous 5-HT, the total areas of TCA aggregates in the PMBSF and those in visual cortex were similar to the controls. These results suggest that TCAs have a graded response to increasingly higher 5-HT concentrations. The lack of TCA expansion beyond normal cortical areas further implies that 5-HT-induced axon outgrowth is restricted at cortical boundaries.
Collapse
Affiliation(s)
- Richard D Lane
- Department of Neurosciences, Medical University of Ohio at Toledo, 3035 Arlington Avenue, Toledo, OH 43614-5804, USA
| | | | | | | | | |
Collapse
|
47
|
He JC, Gomes I, Nguyen T, Jayaram G, Ram PT, Devi LA, Iyengar R. The G alpha(o/i)-coupled cannabinoid receptor-mediated neurite outgrowth involves Rap regulation of Src and Stat3. J Biol Chem 2005; 280:33426-34. [PMID: 16046413 DOI: 10.1074/jbc.m502812200] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The study of the signaling pathways regulating neurite outgrowth in culture is important because of their potential role in neuronal differentiation in vivo. We have previously shown that the G alpha(o/i)-coupled CB1 cannabinoid receptor (CB1R) activates Rap1 to induce neurite outgrowth. G alpha(o/i) also activates the Src-Stat3 pathway. Here, we studied the relationship between the G alpha(o/i)-Rap1 and Src-Stat3 pathways and the role of these signaling pathways in CB1R-mediated neurite outgrowth in Neuro-2A cells. The CB1 agonist HU-210 induced pertussis toxin-sensitive Src and Stat3 phosphorylation. Dominant negative (DN) mutants of Src and Stat3 blocked CB1R-induced neurite outgrowth. Constitutively active Rap 1B and Ral-activated Src and CB1R-induced Src phosphorylation was inhibited by Rap1-DN and Ral-DN, indicating that both Rap1 and Ral mediate downstream signaling from G alpha(o/i) for Src activation. Rap1-activated Ral and Ral-DN blocked Rap-induced Src phosphorylation. G alpha(o)-induced Stat3 activation was blocked by Ral-DN, whereas v-Src-induced Stat3 activation was not inhibited by Ral-DN, indicating that the CB1R, through G alpha(o), mediates the sequential activation of Rap1 to Ral to Src to Stat3 in Neuro-2A cells. Downstream of Src, the CB1R also activated Rac1 and JNK, which enhanced CBR1-mediated Stat3 activation. Rac-DN blocked CB1R-induced activation of JNK. Pharmacological inhibition of JNK blocked Src and CB1R activation of Stat3, indicating that Rac and JNK are also involved in CB1R-mediated neurite outgrowth. Overall, this study demonstrated that G alpha(o/i)-coupled CB1R triggers neurite outgrowth in Neuro-2A through the activation of a signaling network containing two pathways that bifurcate at Src and converge at Stat3.
Collapse
Affiliation(s)
- John Cijiang He
- Department of Pharmacology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Russo A, Pellitteri R, Monaco S, Romeo R, Stanzani S. "In vitro" postnatal expression of 5-HT7 receptors in the rat hypothalamus: an immunohistochemical analysis. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 154:211-6. [PMID: 15707674 DOI: 10.1016/j.devbrainres.2004.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2004] [Revised: 11/08/2004] [Accepted: 11/10/2004] [Indexed: 12/01/2022]
Abstract
The neurotransmitter serotonin (5-HT) is involved in various physiological functions via multiple receptor subtypes. These have been classified in seven receptor families (5-HT1-7) on the basis of their structures and functional characteristics. In this study, we examined the expression of 5-HT7 receptors in the rat hypothalamic neurons cultured "in vitro" during postnatal development. Neuronal cultures were prepared from postnatal pups P2, P3 and P5, were treated with bFGF and processed by means of immunofluorescence technique using a polyclonal 5-HT7 receptor antibody. In P2, we found a low density of 5-HT7 labeled hypothalamic bFGF-treated neurons and no 5-HT7 immunolabeling in control cultures; in P3, a moderate number of bFGF-treated neurons were observed but they were not bright. No 5-HT7 immunolabeling was found in controls. In P5, a heavy labeling of small sized bipolar neurons was seen in bFGF-treated neurons, while in control cultures, few labeled neurons with a low stained density were observed. These results suggest that 5-HT7 receptors in hypothalamic neurons begin to appear at P5 and then could be involved in many physiological implications that are not exerted at P2 and P3. This indicates that 5-HT7 receptors have a potential significance in the rat hypothalamus during early postnatal development.
Collapse
Affiliation(s)
- Antonella Russo
- Department of Physiological Sciences, University of Catania, Viale Andrea Doria 6, Catania 95025, Italy.
| | | | | | | | | |
Collapse
|
49
|
Jordan JD, He JC, Eungdamrong NJ, Gomes I, Ali W, Nguyen T, Bivona TG, Philips MR, Devi LA, Iyengar R. Cannabinoid receptor-induced neurite outgrowth is mediated by Rap1 activation through G(alpha)o/i-triggered proteasomal degradation of Rap1GAPII. J Biol Chem 2005; 280:11413-21. [PMID: 15657046 DOI: 10.1074/jbc.m411521200] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The G(alpha)o/i-coupled CB1 cannabionoid receptor induces neurite outgrowth in Neuro-2A cells. The mechanisms of signaling through G(alpha)o/i to induce neurite outgrowth were studied. The expression of G(alpha)o/i reduces the stability of its direct interactor protein, Rap1GAPII, by targeting it for ubiquitination and proteasomal degradation. This results in the activation of Rap1. G(alpha)o/i-induced activation of endogenous Rap1 in Neuro-2A cells is blocked by the proteasomal inhibitor lactacystin. G(alpha)o/i stimulates neurite outgrowth that is blocked by the expression of dominant negative Rap1. Expression of Rap1GAPII also blocks the G(alpha)o/i-induced neurite outgrowth and treatment with proteasomal inhibitors potentiates this inhibition. The endogenous G(alpha)o/i-coupled cannabinoid (CB1) receptor in Neuro-2A cells stimulates the degradation of Rap1GAPII; activation of Rap1 and treatment with pertussis toxin or lactacystin blocks these effects. The CB1 receptor-stimulated neurite outgrowth is blocked by treatment with pertussis toxin, small interfering RNA for Rap, lactacystin, and expression of Rap1GAPII. Thus, the G(alpha)o/i-coupled cannabinoid receptor, by regulating the proteasomal degradation of Rap1GAPII, activates Rap1 to induce neurite outgrowth.
Collapse
Affiliation(s)
- J Dedrick Jordan
- Department of Pharmacology and Biological Chemistry, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Stanwood GD, Parlaman JP, Levitt P. Anatomical abnormalities in dopaminoceptive regions of the cerebral cortex of dopamine D1 receptor mutant mice. J Comp Neurol 2005; 487:270-82. [PMID: 15892099 DOI: 10.1002/cne.20548] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Alteration of dopamine neurotransmission during development can induce specific changes in neuronal structure and function. Here, we report specific morphological and neurochemical changes of projection neurons and interneurons of the medial frontal cortex of the dopamine D(1) receptor null mouse. Using immunostaining of cytoskeletal proteins and a crossbred D(1) receptor null:YFP transgenic reporter line, we demonstrate that the apical dendrites of pyramidal cells are abnormally organized in the prefrontal and anterior cingulate cortices of mice lacking the D(1) receptor. Neuronal processes exhibit a decrease in bundling and an increase in irregular, tortuous patterning as they weave a course towards the pial surface. In addition, there is increased parvalbumin staining of the dendrites of cortical interneurons in D(1) receptor null mice. Both pyramidal and interneuron alterations are evident by the early postnatal period and persist into adulthood. The alterations show regional specificity, in that dendritic profiles of projection neurons and interneurons in somatosensory and visual cortices develop normally. The abnormalities are reminiscent of those induced by prenatal exposure to cocaine in rabbits, an insult which has been shown to produce an attenuation of D(1) receptor-mediated responses through G(salpha). These results suggest that loss of D(1) receptor-mediated signaling during development produces permanent alterations in the cellular organization of specific cortical areas involved in attention, cognition, and emotion. Pharmacological and behavioral studies in the D(1) null mouse should be interpreted in the context of possible altered circuitry, given the presence of these developmental defects in the organization of dopaminoceptive regions of the cerebral cortex.
Collapse
Affiliation(s)
- Gregg D Stanwood
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37203, USA.
| | | | | |
Collapse
|