1
|
Bellon A. Comparing stem cells, transdifferentiation and brain organoids as tools for psychiatric research. Transl Psychiatry 2024; 14:127. [PMID: 38418498 PMCID: PMC10901833 DOI: 10.1038/s41398-024-02780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 03/01/2024] Open
Abstract
The inaccessibility of neurons coming directly from patients has hindered our understanding of mental illnesses at the cellular level. To overcome this obstacle, six different cellular approaches that carry the genetic vulnerability to psychiatric disorders are currently available: Olfactory Neuroepithelial Cells, Mesenchymal Stem Cells, Pluripotent Monocytes, Induced Pluripotent Stem Cells, Induced Neuronal cells and more recently Brain Organoids. Here we contrast advantages and disadvantages of each of these six cell-based methodologies. Neuronal-like cells derived from pluripotent monocytes are presented in more detail as this technique was recently used in psychiatry for the first time. Among the parameters used for comparison are; accessibility, need for reprograming, time to deliver differentiated cells, differentiation efficiency, reproducibility of results and cost. We provide a timeline on the discovery of these cell-based methodologies, but, our main goal is to assist researchers selecting which cellular approach is best suited for any given project. This manuscript also aims to help readers better interpret results from the published literature. With this goal in mind, we end our work with a discussion about the differences and similarities between cell-based techniques and postmortem research, the only currently available tools that allow the study of mental illness in neurons or neuronal-like cells coming directly from patients.
Collapse
Affiliation(s)
- Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA, USA.
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, USA.
| |
Collapse
|
2
|
McMahon DG, Dowling JE. Neuromodulation: Actions of Dopamine, Retinoic Acid, Nitric Oxide, and Other Substances on Retinal Horizontal Cells. Eye Brain 2023; 15:125-137. [PMID: 37928979 PMCID: PMC10625386 DOI: 10.2147/eb.s420050] [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: 05/05/2023] [Accepted: 08/18/2023] [Indexed: 11/07/2023] Open
Abstract
Whereas excitation and inhibition of neurons are well understood, it is clear that neuromodulatory influences on neurons and their synapses play a major role in shaping neural activity in the brain. Memory and learning, emotional and other complex behaviors, as well as cognitive disorders have all been related to neuromodulatory mechanisms. A number of neuroactive substances including monoamines such as dopamine and neuropeptides have been shown to act as neuromodulators, but other substances thought to play very different roles in the body and brain act as neuromodulators, such as retinoic acid. We still understand little about how neuromodulatory substances exert their effects, and the present review focuses on how two such substances, dopamine and retinoic acid, exert their effects. The emphasis is on the underlying neuromodulatory mechanisms down to the molecular level that allow the second order bipolar cells and the output neurons of the retina, the ganglion cells, to respond to different environmental (ie lighting) conditions. The modulation described affects a simple circuit in the outer retina, involves several neuroactive substances and is surprisingly complex and not fully understood.
Collapse
Affiliation(s)
- Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA
| | - John E Dowling
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
3
|
Bellon A, Feuillet V, Cortez-Resendiz A, Mouaffak F, Kong L, Hong LE, De Godoy L, Jay TM, Hosmalin A, Krebs MO. Dopamine-induced pruning in monocyte-derived-neuronal-like cells (MDNCs) from patients with schizophrenia. Mol Psychiatry 2022; 27:2787-2802. [PMID: 35365810 PMCID: PMC9156413 DOI: 10.1038/s41380-022-01514-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 02/05/2022] [Accepted: 02/25/2022] [Indexed: 01/10/2023]
Abstract
The long lapse between the presumptive origin of schizophrenia (SCZ) during early development and its diagnosis in late adolescence has hindered the study of crucial neurodevelopmental processes directly in living patients. Dopamine, a neurotransmitter consistently associated with the pathophysiology of SCZ, participates in several aspects of brain development including pruning of neuronal extensions. Excessive pruning is considered the cause of the most consistent finding in SCZ, namely decreased brain volume. It is therefore possible that patients with SCZ carry an increased susceptibility to dopamine's pruning effects and that this susceptibility would be more obvious in the early stages of neuronal development when dopamine pruning effects appear to be more prominent. Obtaining developing neurons from living patients is not feasible. Instead, we used Monocyte-Derived-Neuronal-like Cells (MDNCs) as these cells can be generated in only 20 days and deliver reproducible results. In this study, we expanded the number of individuals in whom we tested the reproducibility of MDNCs. We also deepened the characterization of MDNCs by comparing its neurostructure to that of human developing neurons. Moreover, we studied MDNCs from 12 controls and 13 patients with SCZ. Patients' cells differentiate more efficiently, extend longer secondary neurites and grow more primary neurites. In addition, MDNCs from medicated patients expresses less D1R and prune more primary neurites when exposed to dopamine. Haloperidol did not influence our results but the role of other antipsychotics was not examined and thus, needs to be considered as a confounder.
Collapse
Affiliation(s)
- Alfredo Bellon
- Department of Psychiatry and Behavioral Health, Penn State Hershey Medical Center, Hershey, PA, USA.
- Department of Pharmacology, Penn State Hershey Medical Center, Hershey, PA, USA.
| | - Vincent Feuillet
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Alonso Cortez-Resendiz
- Department of Psychiatry and Behavioral Health, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Faycal Mouaffak
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
- Pôle de Psychiatrie d'Adultes 93G04, EPS Ville Evrard, Saint Denis, France
| | - Lan Kong
- Department of Public Health Sciences, Penn State Hershey Medical Center, Hershey, PA, USA
| | - L Elliot Hong
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Therese M Jay
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
| | - Anne Hosmalin
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Marie-Odile Krebs
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
- Groupe-Hospitalo-Universitaire de Paris, Psychiatrie et Neuroscience, Pôle PEPIT, University of Paris, Paris, France
| |
Collapse
|
4
|
|
5
|
Haug MF, Berger M, Gesemann M, Neuhauss SCF. Differential expression of PKCα and -β in the zebrafish retina. Histochem Cell Biol 2019; 151:521-530. [PMID: 30604284 DOI: 10.1007/s00418-018-1764-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2018] [Indexed: 01/08/2023]
Abstract
The retina is a complex neural circuit, which processes and transmits visual information from light perceiving photoreceptors to projecting retinal ganglion cells. Much of the computational power of the retina rests on signal integrating interneurons, such as bipolar cells. Commercially available antibodies against bovine and human conventional protein kinase C (PKC) α and -β are frequently used as markers for retinal ON-bipolar cells in different species, despite the fact that it is not known which bipolar cell subtype(s) they actually label. In zebrafish (Danio rerio) five prkc genes (coding for PKC proteins) have been identified. Their expression has not been systematically determined. While prkcg is not expressed in retinal tissue, the other four prkc (prkcaa, prkcab, prkcba, prkcbb) transcripts were found in different parts of the inner nuclear layer and some as well in the retinal ganglion cell layer. Immunohistochemical analysis in adult zebrafish retina using fluorescent in situ hybridization and PKC antibodies showed an overlapping immunolabeling of ON-bipolar cells that are most likely of the BON s6 and BON s6L or RRod type. However, comparison of transcript expression with immunolabeling, implies that these antibodies are not specific for one single zebrafish conventional PKC, but rather detect a combination of PKC -α and -β variants.
Collapse
Affiliation(s)
- Marion F Haug
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Manuela Berger
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Matthias Gesemann
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Stephan C F Neuhauss
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| |
Collapse
|
6
|
Si L, Pan L, Zhang X, Wang H, Wei C. Evidence that dopamine is involved in neuroendocrine regulation, gill intracellular signaling pathways and ion regulation in Litopenaeus vannamei. J Exp Biol 2019; 222:jeb.204073. [DOI: 10.1242/jeb.204073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/27/2019] [Indexed: 01/24/2023]
Abstract
The transport of ions and ammonia in the gills may be regulated by neuroendocrine factors, in order to explore the regulation mechanism of dopamine (DA), hemolymph neuroendocrine hormones, gill intracellular signaling pathways, ion and ammonia transporters, as well as hemolymph osmolality and ammonia concentration were investigated in Litopenaeus vannamei after 10−7 and 10−6 mol shrimp−1 DA injection. The data displayed a significant increase in crustacean hyperglycemic hormone (CHH) concentration at 1-12 h and a transient significant decrease in corticotrophin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH) and cortisol concentrations under DA stimulation. The up-regulation of guanylyl cyclase (GC) mRNA, cyclic guanosine monophosphate (cGMP) and protein kinase G (PKG) concentrations, together with down-regulation of DA receptor D4 mRNA and up-regulation of cyclic adenosine monophosphate (cAMP), protein kinase A (PKA), diacylglycerol (DAG) and protein kinase C (PKC) concentrations suggested an activation of complicated intracellular signaling pathway. The expression of cyclic AMP response element-binding protein (CREB), FXYD2 and 14-3-3 protein mRNA was significantly increased by PKA regulation. The increase in Na+/K+-ATPase (NKA) activity and the stabilization of V-type H+-ATPase (V-ATPase) activity are accompanied by an up-regulation of K+-channel, Na+/K+/2Cl− cotransporter (NKCC), Rh protein and vesicle associated membrane protein (VAMP) mRNA, resulting in an increase in hemolymph osmolality and a decrease in hemolymph ammonia concentration. These results suggest that DA stimulates the secretion of CHH and inhibits the release of cortisol, which activates intracellular signaling factors to facilitate ion and ammonia transport across the gills, and may not affect intracellular acidification.
Collapse
Affiliation(s)
- Lingjun Si
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Luqing Pan
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Xin Zhang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Hongdan Wang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| | - Cun Wei
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao 266003, PR China
| |
Collapse
|
7
|
Bellon A, Wegener A, Lescallette AR, Valente M, Yang SK, Gardette R, Matricon J, Mouaffak F, Watts P, Vimeux L, Yun JK, Kawasawa YI, Clawson GA, Blandin E, Chaumette B, Jay TM, Krebs MO, Feuillet V, Hosmalin A. Transdifferentiation of Human Circulating Monocytes Into Neuronal-Like Cells in 20 Days and Without Reprograming. Front Mol Neurosci 2018; 11:323. [PMID: 30760979 PMCID: PMC6156467 DOI: 10.3389/fnmol.2018.00323] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/21/2018] [Indexed: 12/19/2022] Open
Abstract
Despite progress, our understanding of psychiatric and neurological illnesses remains poor, at least in part due to the inability to access neurons directly from patients. Currently, there are in vitro models available but significant work remains, including the search for a less invasive, inexpensive and rapid method to obtain neuronal-like cells with the capacity to deliver reproducible results. Here, we present a new protocol to transdifferentiate human circulating monocytes into neuronal-like cells in 20 days and without the need for viral insertion or reprograming. We have thoroughly characterized these monocyte-derived-neuronal-like cells (MDNCs) through various approaches including immunofluorescence (IF), flow cytometry, qRT-PCR, single cell mRNA sequencing, electrophysiology and pharmacological techniques. These MDNCs resembled human neurons early in development, expressed a variety of neuroprogenitor and neuronal genes as well as several neuroprogenitor and neuronal proteins and also presented electrical activity. In addition, when these neuronal-like cells were exposed to either dopamine or colchicine, they responded similarly to neurons by retracting their neuronal arborizations. More importantly, MDNCs exhibited reproducible differentiation rates, arborizations and expression of dopamine 1 receptors (DR1) on separate sequential samples from the same individual. Differentiation efficiency measured by cell morphology was on average 11.9 ± 1.4% (mean, SEM, n = 38,819 cells from 15 donors). To provide context and help researchers decide which in vitro model of neuronal development is best suited to address their scientific question,we compared our results with those of other in vitro models currently available and exposed advantages and disadvantages of each paradigm.
Collapse
Affiliation(s)
- Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry, Hershey, PA, United States.,Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, United States.,INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France.,Centre Hospitalier Sainte-Anne, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire-S14, Paris, France
| | - Amelie Wegener
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Adam R Lescallette
- Penn State Hershey Medical Center, Department of Psychiatry, Hershey, PA, United States
| | - Michael Valente
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Seung-Kwon Yang
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Robert Gardette
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Julien Matricon
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Faycal Mouaffak
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France.,Centre Hospitalier Sainte-Anne, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire-S14, Paris, France
| | - Paula Watts
- Sky Ridge Medical Center, Department of Internal Medicine, Lone Tree, CO, United States
| | - Lene Vimeux
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Jong K Yun
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, United States.,Penn State Hershey Medical Center, Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Hershey, PA, United States
| | - Gary A Clawson
- Gittlen Cancer Research Laboratories, Department of Pathology, Penn State University College of Medicine, Hershey, PA, United States
| | - Elisabeta Blandin
- Penn State Hershey Medical Center, Department of Psychiatry, Hershey, PA, United States.,Penn State Hershey Medical Center, Neural & Behavioral Sciences, Hershey, PA, United States
| | - Boris Chaumette
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France.,Centre Hospitalier Sainte-Anne, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire-S14, Paris, France.,Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Therese M Jay
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France
| | - Marie-Odile Krebs
- Université Paris Descartes, Sorbonne Paris Cite, Paris, France.,INSERM UMR894, Center for Psychiatry and Neurosciences, Paris, France.,Centre Hospitalier Sainte-Anne, Faculté de Médecine Paris Descartes, Service Hospitalo-Universitaire-S14, Paris, France
| | - Vincent Feuillet
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Anne Hosmalin
- INSERM U1016, Institut Cochin, Paris, France.,CNRS UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cite, Paris, France
| |
Collapse
|
8
|
Haug MF, Gesemann M, Berger M, Neuhauss SCF. Phylogeny and distribution of protein kinase C variants in the zebrafish. J Comp Neurol 2018; 526:1097-1109. [DOI: 10.1002/cne.24395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Marion F. Haug
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology; Zurich CH-8057 Switzerland
| | - Matthias Gesemann
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology; Zurich CH-8057 Switzerland
| | - Manuela Berger
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology; Zurich CH-8057 Switzerland
| | - Stephan C. F. Neuhauss
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology; Zurich CH-8057 Switzerland
| |
Collapse
|
9
|
Abstract
Nervous system development depends on axonal growth cone recognition of extracellular guidance clues and transduction of this information into directed growth. Major advances have been made in characterizing the extracellular molecules that serve as signals for growing axons, in correlating fluctuations of Ca,++with motility, and in demonstrating the actin-dependent basis of growth cone motility. The intracellular events that immediately follow ligand-receptor interaction at the growth cone are largely undetermined. Molecules of the integrin family, the cadherin family, and the cell adhesion molecule family organize cytoskeletal changes directly but also may initiate signaling cascades involving diffusible messengers. Heterotrimeric G proteins are highly concentrated in the growth cone membrane and can account for the initial steps in signal transduction for several neurotransmitters that regulate axonal growth. GAP-43 enhances the sensitivity of G protein-mediated transduction. Molecules inhibitory for neuronal growth, such as collapsin, initiate a signal transduction cascade likely to involve G proteins and an intracellular protein, CRMP-62. Further analysis of growth cone signal transduction will provide a molecular understanding of the development of synaptic connectivity during brain development. The Neuroscientist 2:83-86, 1996
Collapse
|
10
|
Li F, Ohtani A, Senzaki K, Shiga T. Receptor-dependent regulation of dendrite formation of noradrenaline and dopamine in non-GABAergic cerebral cortical neurons. Dev Neurobiol 2012; 73:370-83. [PMID: 23135899 DOI: 10.1002/dneu.22065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 09/18/2012] [Accepted: 10/31/2012] [Indexed: 12/12/2022]
Abstract
The present study characterized the receptor-dependent regulation of dendrite formation of noradrenaline (NA) and dopamine (DA) in cultured neurons obtained from embryonic day 16 rat cerebral cortex. Morphological diversity of cortical dendrites was analyzed on various features: dendrite initiation, dendrite outgrowth, and dendrite branching. Using a combination of immunocytochemical markers of dendrites and GABAergic neurons, we focused on the dendrite morphology of non-GABAergic neurons. Our results showed that (1) NA inhibited the dendrite branching, (2) β adrenergic receptor (β-AR) agonist inhibited the dendrite initiation, while promoted the dendrite outgrowth, (3) β1-AR and β2-AR were present in all the cultured neurons, and both agonists inhibited the dendrite initiation, while only β1-AR agonist induced the dendrite branching; (4) DA inhibited the dendrite outgrowth, (5) D1 receptor agonist inhibited the dendrite initiation, while promoted the dendrite branching. In conclusion, this study compared the effects of NA, DA and their receptors and showed that NA and DA regulate different features on the dendrite formation of non-GABAergic cortical neurons, depending on the receptors.
Collapse
Affiliation(s)
- Fei Li
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Ibaraki, Japan
| | | | | | | |
Collapse
|
11
|
Wu CL, Chou YH, Chang YJ, Teng NY, Hsu HL, Chen L. Interplay between cell migration and neurite outgrowth determines SH2B1β-enhanced neurite regeneration of differentiated PC12 cells. PLoS One 2012; 7:e34999. [PMID: 22539954 PMCID: PMC3335126 DOI: 10.1371/journal.pone.0034999] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 03/08/2012] [Indexed: 11/19/2022] Open
Abstract
The regulation of neurite outgrowth is crucial in developing strategies to promote neurite regeneration after nerve injury and in degenerative diseases. In this study, we demonstrate that overexpression of an adaptor/scaffolding protein SH2B1β promotes neurite re-growth of differentiated PC12 cells, an established neuronal model, using wound healing (scraping) assays. Cell migration and the subsequent remodeling are crucial determinants during neurite regeneration. We provide evidence suggesting that overexpressing SH2B1β enhances protein kinase C (PKC)-dependent cell migration and phosphatidylinositol 3-kinase (PI3K)-AKT-, mitogen activated protein kinase (MAPK)/extracellular signal-regulated protein kinase (ERK) kinase (MEK)-ERK-dependent neurite re-growth. Our results further reveal a cross-talk between pathways involving PKC and ERK1/2 in regulating neurite re-growth and cell migration. We conclude that temporal regulation of cell migration and neurite outgrowth by SH2B1β contributes to the enhanced regeneration of differentiated PC12 cells.
Collapse
Affiliation(s)
- Chia-Ling Wu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Yu-Han Chou
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Yu-Jung Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Nan-Yuan Teng
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Hsin-Ling Hsu
- Division of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan, Republic of China
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
12
|
Ogilvie JM, Hakenewerth AM, Gardner RR, Martak JG, Maggio VM. Dopamine receptor loss of function is not protective of rd1 rod photoreceptors in vivo. Mol Vis 2009; 15:2868-78. [PMID: 20038975 PMCID: PMC2797042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 12/20/2009] [Indexed: 11/16/2022] Open
Abstract
PURPOSE The retinal degeneration (rd1) mouse undergoes a rapid loss of rod photoreceptors due to a defect in the cGMP-phosphodiesterase gene. We have previously demonstrated that dopamine (DA) antagonists or DA depletion blocks photoreceptor degeneration and that DA is necessary for photoreceptor degeneration in the rd1 mouse retinal organ culture model. Antagonists for either D1- or D2-family DA receptors are protective in rd1 organ cultures. METHODS To determine whether photoreceptor survival can be increased in vivo in the rd1 mouse, we used both a pharmacological and a genetic approach. The pharmacological approach involved three techniques to administer 6-hydroxydopamine (6-OHDA) in an attempt to deplete DA in postnatal mouse retina in vivo. As a genetic alternative, DA receptor signaling was inactivated by crossbreeding rd1 mice to D1, D2, D4, and D5 knockout mice to create four lines of double mutants. RESULTS Pharmacological DA depletion was incomplete due to the limiting size of the postnatal mouse eye and the lethality of systemic inhibition of DA signaling. In all four lines of double mutants, no increase in rod photoreceptor survival was observed. To determine whether protection of rd1 photoreceptors by inhibition of dopaminergic signaling is a result of conditions specific to the organ culture environment, we grew in vitro retinas from the four lines of double mutant mice for four weeks. Again, no increase in photoreceptor survival was seen. Finally, three triple mutants were generated that lacked two DA receptors (D1/D2; D1/D4; and D2/D4) on a rd1 background. In all three cases, rod photoreceptors were not protected from degeneration. CONCLUSIONS The dramatic protection of rd1 rod photoreceptors by inhibition of DA signaling in organ culture has not been reproduced in vivo by either a pharmacological approach, due to technical limitations, or by genetic manipulations. The possible role of compensatory effects during retinal development in DA receptor deficient mice is considered.
Collapse
|
13
|
Saito M, Toyoda H, Sato H, Ishii H, Kang Y. Rapid use-dependent down-regulation of γ-aminobutyric acid type A receptors in rat mesencephalic trigeminal neurons. J Neurosci Res 2009; 87:3120-33. [DOI: 10.1002/jnr.22141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
14
|
Reis RAM, Ventura ALM, Kubrusly RCC, de Mello MCF, de Mello FG. Dopaminergic signaling in the developing retina. ACTA ACUST UNITED AC 2007; 54:181-8. [PMID: 17292477 DOI: 10.1016/j.brainresrev.2007.01.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 01/03/2007] [Accepted: 01/09/2007] [Indexed: 11/16/2022]
Abstract
The role of dopamine in the retina has been studied for the last 30 years and there is now increasing evidence that dopamine is used as a developmental signal in the embryonic retina. Dopamine is the main catecholamine found in the retina of most species, being synthesized from the L-amino acid tyrosine. Its effects are mediated by G protein coupled receptors constituting the D(1) (D(1) and D(5)) and D(2) (D(2), D(3) and D(4)) receptor subfamilies that can be coupled to adenylyl cyclase in opposite manners. Dopamine-mediated cyclic AMP (cAMP) accumulation, via D(1)-like receptors, is observed very early during retina ontogeny, before synaptogenesis and, in some species, before the expression of tyrosine hydroxylase (TH), the enzyme that characterizes the neuronal dopaminergic phenotype. D(2)-like receptors appear in the tissue days after D(1)-like activity is detected. In the embryonic avian retina, before the tissue is capable of synthesizing its own dopamine via TH, dopamine synthesis is observed from L-DOPA supplied to the neuroretina from retina pigmented epithelium which results in dopaminergic communication in the embryonic tissue before TH expression. Müller cells, the main glia type found in the retina, seem to actively contribute to dopaminergic activity in the retinal tissue. Understanding the dopaminergic role during retina development may contribute to novel strategies approaching certain visual dysfunctions such as those found in ocular albinism.
Collapse
Affiliation(s)
- Ricardo A M Reis
- Lab. Neurochemistry, Program in Neurobiology IBCCF, UFRJ, Rio de Janeiro, Brazil.
| | | | | | | | | |
Collapse
|
15
|
Tyler MJ, Cameron DA. Cellular pattern formation during retinal regeneration: a role for homotypic control of cell fate acquisition. Vision Res 2006; 47:501-11. [PMID: 17034830 DOI: 10.1016/j.visres.2006.08.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 08/23/2006] [Accepted: 08/24/2006] [Indexed: 11/20/2022]
Abstract
A dominant mechanism of cellular patterning in the growing fish retina is control of cell fate acquisition by negative feedback signals arising from differentiated cells. We tested the ability of a computational model of this pattern formation mechanism to simulate cellular patterns in regenerated goldfish retina. The model successfully simulated quantitative features of in vivo regenerated patterns, indicating that regenerating retina has access to and utilizes patterning mechanisms that are operational during normal growth. The atypical patterns of regenerated retina could arise in part from regenerative progenitors that, compared to normal growth progenitors, are less responsive to the feedback patterning signals.
Collapse
Affiliation(s)
- Melinda J Tyler
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY 13210, USA
| | | |
Collapse
|
16
|
Tyler MJ, Carney LH, Cameron DA. Control of cellular pattern formation in the vertebrate inner retina by homotypic regulation of cell-fate decisions. J Neurosci 2006; 25:4565-76. [PMID: 15872104 PMCID: PMC6725033 DOI: 10.1523/jneurosci.0588-05.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The vertebrate retina is composed of cellular arrays that are nonrandom across two-dimensional space. The determinants of these nonrandom two-dimensional cellular patterns in the inner nuclear layer of the retina were investigated using empirical and computational modeling techniques. In normal and experimental models of goldfish retinal growth, the patterns of tyrosine hydroxylase- and serotonin-positive cells indicated that neither cell death nor lateral migration of differentiated cells were dominant mechanisms of cellular pattern formation. A computational model of cellular pattern formation that used a signaling mechanism arising from differentiated cells that inhibited homotypic cell-fate decisions generated accurate simulations of the empirically observed patterns in normal retina. This model also predicted the principal atypical cellular pattern characteristic, a transient cell-type-specific hyperplasia, which was empirically observed in the growing retina subsequent to selective ablation of differentiated retinal cells, either tyrosine hydroxylase positive or serotonin positive. The results support the hypothesis that inhibitory spatiotemporal regulation of homotypic cell-fate decisions is a dominant mechanistic determinant of nonrandom cellular patterns in the vertebrate retina.
Collapse
Affiliation(s)
- Melinda J Tyler
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York 13210, USA
| | | | | |
Collapse
|
17
|
FRIEDMAN EITAN, WANG HOAUYAN. Prenatal Cocaine Exposure Alters Signal Transduction in the Brain D1Dopamine Receptor Systema. Ann N Y Acad Sci 2006; 846:238-247. [DOI: 10.1111/j.1749-6632.1998.tb09741.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Larsson C. Protein kinase C and the regulation of the actin cytoskeleton. Cell Signal 2005; 18:276-84. [PMID: 16109477 DOI: 10.1016/j.cellsig.2005.07.010] [Citation(s) in RCA: 290] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Revised: 07/18/2005] [Accepted: 07/18/2005] [Indexed: 01/01/2023]
Abstract
Protein kinase C (PKC) isoforms are central components in intracellular networks that regulate a vast number of cellular processes. It has long been known that in most cell types, one or more PKC isoforms influences the morphology of the F-actin cytoskeleton and thereby regulates processes that are affected by remodelling of the microfilaments. These include cellular migration and neurite outgrowth. This review focuses on the role of classical and novel PKC isoforms in migration and neurite outgrowth, and highlights some regulatory steps that may be of importance in the regulation by PKC of migration and neurite outgrowth. Many studies indicate that integrins are crucial mediators both upstream and downstream of PKC in inducing morphological changes. Furthermore, a number of PKC substrates, directly associated with the microfilaments, such as MARCKS, GAP43, adducin, fascin, ERM proteins and others have been identified. Their potential role in PKC effects on the cytoskeleton is discussed.
Collapse
Affiliation(s)
- Christer Larsson
- Lund University, Dept of Laboratory Medicine, Molecular Medicine, Entrance 78, 3rd floor, UMAS SE-205 02, Malmö University Hospital, Malmö, Sweden.
| |
Collapse
|
19
|
Abstract
The present paper reports on the results obtained in a rabbit model of prenatal cocaine exposure that mimics the pharmacokinetics of crack cocaine in humans, and relates these findings to studies in other species including humans. A general finding is that prenatal exposure to cocaine during neurogenesis produces dysfunctions in signal transduction via the dopamine D(1) receptor and alterations in cortical neuronal development leading to permanent morphological abnormalities in frontocingulate cortex and other brain structures. Differences in the precise effects obtained appear to be due to the dose, route and time of cocaine administration. Related to these effects of in utero cocaine exposure, animals demonstrate permanent deficits in cognitive processes related to attentional focus that have been correlated with impairment of stimulus processing in the anterior cingulate cortex. The long-term cognitive deficits observed in various species are in agreement with recent reports indicating that persistent attentional and other cognitive deficits are evident in cocaine-exposed children as they grow older and are challenged to master more complex cognitive tasks.
Collapse
Affiliation(s)
- John A Harvey
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia PA 19102-1192, USA.
| |
Collapse
|
20
|
Unterwald EM, Ivkovic S, Cuntapay M, Stroppolo A, Guinea B, Ehrlich ME. Prenatal exposure to cocaine decreases adenylyl cyclase activity in embryonic mouse striatum. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 147:67-75. [PMID: 14741752 DOI: 10.1016/s0165-3806(03)00058-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Adenylyl cyclase activity was measured in the striatum of naive mice as a function of age and in mice exposed in utero to cocaine. In naive Swiss-Webster mice, basal and forskolin-stimulated adenylyl cyclase activity increased gradually from embryonic day 13 (E13) until 2-3 weeks of age when activity peaked before decreasing slightly to adult levels. The ability of the dopamine D1 receptor agonist, SKF 82958, to stimulate adenylyl cyclase activity also increased in magnitude until P15. In a separate study, pregnant Swiss-Webster mice were injected twice daily with cocaine (15 mg/kg, s.c.) or an equal volume of saline from E10 to E17. Adenylyl cyclase activity was measured in the striatum of E18 embryos. Basal adenylyl cyclase activity was significantly reduced following prenatal exposure to cocaine. Likewise, the ability of forskolin or SKF 82958 to stimulate adenylyl cyclase was attenuated following cocaine exposure. DeltaFosB was not induced, contrary to what is seen in adult mice. These results demonstrate a functional change in a critical signal transduction pathway following chronic in utero exposure to cocaine that might have profound effects of the development of the brain. Alterations in the cAMP system may underlie some of the deficits seen in humans exposed in utero to cocaine.
Collapse
Affiliation(s)
- Ellen M Unterwald
- Department of Pharmacology, Temple University School of Medicine, 3420 N. Broad St., Philadelphia, PA 19140, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Kurshan PT, Hamilton IS, Mustard JA, Mercer AR. Developmental changes in expression patterns of two dopamine receptor genes in mushroom bodies of the honeybee,Apis mellifera. J Comp Neurol 2003; 466:91-103. [PMID: 14515242 DOI: 10.1002/cne.10864] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The expression patterns of two dopamine receptor genes, Amdop1 and Amdop2, in the developing mushroom bodies of the honeybee brain were determined by using in situ hybridisation. Both genes were expressed throughout pupal development, but their patterns of expression in the three major divisions of mushroom body intrinsic neurons (outer compact cells, noncompact cells, and inner compact cells) were quite distinct. Amdop1 expression could be detected in all three mushroom body cell groups throughout development. Staining for Amdop1 mRNA was particularly intense in newly born Kenyon cells, suggesting that levels of Amdop1 expression are higher in newborn cells than in more mature mushroom body neurons. This was not the case for Amdop2. Amdop2 expression in the mushroom bodies was restricted to inner and outer compact cells during most of pupal development, appearing in noncompact cells only late in metamorphosis or at adult eclosion. In contrast to the case with Amdop1, staining for Amdop2 mRNA was observed in glial cells. Expression of Amdop2 in glial cells was detected only at early stages of glial cell development, when the cells are reported to be actively dividing. This study not only implicates dopamine in the development of honeybee mushroom bodies but also suggests different roles for the two dopamine receptors investigated.
Collapse
Affiliation(s)
- Peri T Kurshan
- Department of Zoology, University of Otago, Dunedin, New Zealand
| | | | | | | |
Collapse
|
22
|
Bergson C, Levenson R, Goldman-Rakic PS, Lidow MS. Dopamine receptor-interacting proteins: the Ca(2+) connection in dopamine signaling. Trends Pharmacol Sci 2003; 24:486-92. [PMID: 12967774 DOI: 10.1016/s0165-6147(03)00232-3] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Abnormal activity of the dopamine system has been implicated in several psychiatric and neurological illnesses; however, lack of knowledge about the precise sites of dopamine dysfunction has compromised our ability to improve the efficacy and safety of dopamine-related drugs used in treatment modalities. Recent work suggests that dopamine transmission is regulated via the concerted efforts of a cohort of cytoskeletal, adaptor and signaling proteins called dopamine receptor-interacting proteins (DRIPs). The discovery that two DRIPs, calcyon and neuronal Ca(2+) sensor 1 (NCS-1), are upregulated in schizophrenia highlights the possibility that altered protein interactions and defects in Ca(2+) homeostasis might contribute to abnormalities in the brain dopamine system in neuropsychiatric diseases.
Collapse
Affiliation(s)
- Clare Bergson
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | | | |
Collapse
|
23
|
Kim BJ, Ghil SH, Kim MJ, Yun Park S, Kim DS, Hwan Kim S, Chin H, Birnbaumer L, Jiang M, Hong SY, Suh-Kim H, Lee YD. Modulation of the N-type calcium channel gene expression by the alpha subunit of Go. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 112:95-102. [PMID: 12670707 DOI: 10.1016/s0169-328x(03)00053-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Go, a heterotrimeric G-protein, is enriched in brain and neuronal growth cones. Although several reports suggest that Go may be involved in modulation of neuronal differentiation, the precise role of Go is not clear. To investigate the function of Go in neuronal differentiation, we determined the effect of Goalpha, the alpha subunit of Go, on the expression of Ca(v)2.2, the pore-forming unit of N-type calcium channels, at the transcription level. Treatment with cyclic AMP (cAMP), which triggers neurite outgrowth in neuroblastoma F11 cells, increased the mRNA level and the promoter activity of the Ca(v)2.2 gene. Overexpression of Goalpha inhibited neurite extension in F11 cells and simultaneously repressed the stimulatory effect of cAMP on the Ca(v)2.2 gene expression to the basal level. Targeted mutation of the Goalpha gene also increased the level of Ca(v)2.2 in the brain. These results suggest that Go may regulate neuronal differentiation through modulation of gene expression of target genes such as N-type calcium channels.
Collapse
Affiliation(s)
- Bum-Jun Kim
- Department of Anatomy, School of Medicine, Ajou University, San 5, Wonchon-dong, Paldal-gu, Suwon, 442-749, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Sacca GB, Sáenz DA, Jaliffa CO, Minces L, Keller Sarmiento MI, Rosenstein RE. Photic regulation of heme oxygenase activity in the golden hamster retina: involvement of dopamine. J Neurochem 2003; 85:534-42. [PMID: 12675930 DOI: 10.1046/j.1471-4159.2003.01697.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The photic regulation of heme oxygenase (HO) activity was examined in the golden hamster retina. This enzymatic activity was significantly higher at midday than at midnight. When the hamsters were placed under constant darkness for 48 h and killed at subjective day or at subjective night, the differences in HO activity disappeared. Western blot analysis showed no differences in HO levels among these time points. Dopamine significantly increased this activity in retinas excised at noon or at midnight, with a higher sensitivity at night. The effect of dopamine was reversed by SCH 23390 but not by spiperone and clozapine and it was not reproduced by quinpirole. In vitro, the increase in HO activity found in retinas incubated under light for 1 h was significantly reduced by SCH 23390. Two cAMP analogs increased HO activity and their effect, as well as the effect of dopamine was blocked by H-89, a protein kinase A (PKA) inhibitor. Tin protoporphyrin IX, an HO inhibitor, significantly decreased cGMP accumulation with maximal effects during the day. Low concentrations of bilirubin decreased retinal thiobarbituric acid substances levels (an index of lipid peroxidation) in basal conditions and after exposing retinal cells to H2O2. These results suggest that hamster retinal HO activity is regulated by the photic stimulus, probably through a dopamine/cAMP/PKA dependent pathway.
Collapse
Affiliation(s)
- Geraldine B Sacca
- Laboratorio de Neuroquímica Retiniana y Oftalmología Experimental, Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
25
|
D1 dopamine receptor regulation of microtubule-associated protein-2 phosphorylation in developing cerebral cortical neurons. J Neurosci 2002. [PMID: 12122070 DOI: 10.1523/jneurosci.22-14-06092.2002] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This study addresses the hypothesis that the previously described capacity of D1 dopamine receptors (D1Rs) to regulate dendritic growth in developing cortical neurons may involve alterations in the phosphorylation state of microtubule-associated protein-2 (MAP2). The changes in phosphorylation of this protein are known to affect its ability to stabilize the dendritic cytoskeleton. The study involved two systems: primary cultures of mouse cortical neurons grown in the presence of the D1R agonists, SKF82958 or A77636, and the cortex of neonatal transgenic mice overexpressing the D1A subtype of D1R. In both models, a decrease in dendritic extension corresponded with an elevation in MAP2 phosphorylation. This phosphorylation occurred on all three amino acid residues examined in this study: serine, threonine, and tyrosine. In cultured cortical neurons, D1R stimulation-induced increase in MAP2 phosphorylation was blocked by the protein kinase A (PKA) inhibitor, H-89, and mimicked by the PKA activator, S(p)-cAMPS. This indicates that D1Rs modulate MAP2 phosphorylation through PKA-associated intracellular signaling pathways. We also observed that the elevations in MAP2 phosphorylation in neuronal cultures in the presence of D1R agonists (or S(p)-cAMPS) were maintained for a prolonged time (up to at least 96 hr). Moreover, MAP2 phosphorylation underwent a substantial increase between 24 and 72 hr of exposure to these drugs. Our findings are consistent with the idea that D1Rs can modulate growth and maintenance of dendrites in developing cortical cells by regulating the phosphorylation of MAP2. In addition, our observations suggest that MAP2 phosphorylation by long-term activation of D1Rs (and PKA) can be divided into two phases: the initial approximately 24-hr-long phase of a relatively weak elevation in phosphorylation and the delayed phase of a much more robust phosphorylation increase taking place during the next approximately 48 hr.
Collapse
|
26
|
Akopian A, McReynolds J, Weiler R. Activation of Protein Kinase C Modulates Light Responses in Horizontal Cells of the Turtle Retina. Eur J Neurosci 2002; 4:745-749. [PMID: 12106318 DOI: 10.1111/j.1460-9568.1992.tb00183.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The effect of phorbol esters on the light-evoked responses of horizontal cells were studied in the turtle eyecup preparation. Phorbol esters caused a reduction in receptive field size and a significant decrease in the amplitude of responses to annular and full-field illumination; however, they caused only minor changes in responses to small spots in the receptive field centre. The dark membrane potential was not affected. The results suggest that phorbol esters may affect both coupling resistance and membrane resistance in horizontal cells. The effects of phorbol esters were blocked by the protein kinase C inhibitor staurosporine, and inactive phorbol ester had no effect, making it very likely that the phorbol ester effects were mediated through activation of protein kinase C. The above effects of the phorbol esters were considerably reduced by the dopamine antagonists haloperidol and fluphenazine, suggesting that they were in part mediated by release of dopamine.
Collapse
Affiliation(s)
- Abram Akopian
- Department of Neurobiology, University of Oldenburg, PO Box 2503, 2900 Oldenburg, FRG
| | | | | |
Collapse
|
27
|
Ogilvie JM, Speck JD. Dopamine has a critical role in photoreceptor degeneration in the rd mouse. Neurobiol Dis 2002; 10:33-40. [PMID: 12079402 DOI: 10.1006/nbdi.2002.0489] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Photoreceptors receive paracrine input from dopaminergic interplexiform cells. Rod photoreceptors in the rd mouse degenerate rapidly due to a specific gene defect. We investigated the effects of dopamine on rd mouse photoreceptors in retinal organ culture. Retinas were harvested from rd or wild-type mice at postnatal day 2 and grown in organ culture for 27 days. When antagonists for either D(1)- or D(2)-family dopamine receptors were added to the media, photoreceptor degeneration was blocked. Furthermore, when dopamine was depleted by the addition of 6-hydroxydopamine and pargyline, photoreceptor survival appeared comparable to wild-type retinal cultures. The addition of a dopamine agonist induced photoreceptor degeneration in dopamine-depleted rd organ cultures. In all cases, photoreceptors maintained robust staining of opsin. These results demonstrate that dopamine antagonists or dopamine depletion blocks photoreceptor degeneration and that dopamine is necessary for photoreceptor degeneration in the rd mouse retinal organ culture model, indicating that dopamine antagonists may represent a therapeutic strategy in retinal degenerative disease.
Collapse
Affiliation(s)
- Judith Mosinger Ogilvie
- Faye and Carl Simons Center for Research in Hearing and Deafness, Central Institute for the Deaf, St. Louis, Missouri, USA
| | | |
Collapse
|
28
|
Abstract
This paper reviews the evidence that antipsychotic drugs induce neuroplasticity. We outline how the synaptic changes induced by the antipsychotic drug haloperidol may help our understanding of the mechanism of action of antipsychotic drugs in general, and how they may help to elucidate the neurobiology of schizophrenia. Studies have provided compelling evidence that haloperidol induces anatomical and molecular changes in the striatum. Anatomical changes have been documented at the level of regional brain volume, synapse morphology, and synapse number. At the molecular level, haloperidol has been shown to cause phosphorylation of proteins and to induce gene expression. The molecular responses to conventional antipsychotic drugs are predominantly observed in the striatum and nucleus accumbens, whereas atypical antipsychotic drugs have a subtler and more widespread impact. We conclude that the ability of antipsychotic drugs to induce anatomical and molecular changes in the brain may be relevant for their antipsychotic properties. The delayed therapeutic action of antipsychotic drugs, together with their promotion of neuroplasticity suggests that modification of synaptic connections by antipsychotic drugs is important for their mode of action. The concept of schizophrenia as a disorder of synaptic organization will benefit from a better understanding of the synaptic changes induced by antipsychotic drugs.
Collapse
Affiliation(s)
- C Konradi
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
29
|
Maharajan P, Maharajan V, Ravagnan G, Paino G. The weaver mutant mouse: a model to study the ontogeny of dopamine transmission systems and their role in drug addiction. Prog Neurobiol 2001; 64:269-76. [PMID: 11240309 DOI: 10.1016/s0301-0082(00)00061-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dopaminergic neurons and their projection-systems are important in some fundamental human activities like locomotion, feeding and sex, essential for survival and procreation, and are relevant to pathologies like Parkinson's disease and drug abuse. Three main dopaminergic projection-systems, namely the nigrostriatal, mesocortical and mesolimbic pathways are the major targets of the neuropharmacological actions of psychomotor stimulants such as cocaine and amphetamine. Studies on knockout mice for dopamine or its receptors provide substantial information but fail to reveal the role of individual dopaminergic projection-systems. Mutant animals with defects specific to one or more projection-systems might be useful for studying the role of individual dopaminergic projection-systems. We propose the weaver mutant mouse, with a defective nigrostriatal dopaminergic projection-system and dopamine depletion in the dorsal striatum but with intact mesocorticolimbic projection-systems, as a suitable model to study the role of individual dopaminergic systems in diverse biological processes including Parkinson's disease and drug abuse.
Collapse
Affiliation(s)
- P Maharajan
- CNR Institute of Cybernetics, 80072 Arco Felice, Naples, Italy
| | | | | | | |
Collapse
|
30
|
Harvey JA, Romano AG, Gabriel M, Simansky KJ, Du W, Aloyo VJ, Friedman E. Effects of prenatal exposure to cocaine on the developing brain: anatomical, chemical, physiological and behavioral consequences. Neurotox Res 2001; 3:117-43. [PMID: 15111265 DOI: 10.1007/bf03033234] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Earlier studies of human infants and studies employing animal models had indicated that prenatal exposure to cocaine produced developmental changes in the behavior of the offspring. The present paper reports on the results obtained in a rabbit model of in utero exposure to cocaine using intravenous injections (4 mg/kg, twice daily) that mimic the pharmacokinetics of crack cocaine in humans. At this dose, cocaine had no effect on the body weight gain of dams, time to delivery, litter size and body weight or other physical characteristics of the offspring. In spite of an otherwise normal appearance, cocaine-exposed neonates displayed a permanent impairment in signal transduction via the D1 dopamine receptor in caudate nucleus, frontal cortex and cingulate cortex due to an uncoupling of the receptor from its associated Gs protein. This uncoupling in the caudate nucleus was shown to have behavioral consequences in that young or adult rabbits, exposed to cocaine in utero, failed to demonstrate amphetamine-elicited motor responses normally seen after activation of D1 receptors in the caudate. The cocaine progeny also demonstrated permanent morphological abnormalities in the anterior cingulate cortex due to uncoupling of the D1 receptor and the consequent inability of dopamine to regulate neurite outgrowth during neuronal development. Consistent with the known functions of the anterior cingulate cortex, adult cocaine progeny demonstrated deficits in attentional processes. This was reflected by impairment in discrimination learning during classical conditioning that was due to an inability to ignore salient stimuli even when these were not relevant to the task. The impairment in discrimination learning also occurred in an instrumental avoidance task and could be shown to be due to an impairment of cingulothalamic learning-related neuronal coding. It was proposed that the selective loss of D1-related neurotransmission in the anterior cingulate cortex prevented an appropriate activation of GABA neurons and thus a loss of inhibitory regulation that is necessary for processes involved in associative attention. Taken together, these findings suggest that the uncoupling of the D1 receptor from its G protein may be the fundamental source of the anatomic, cognitive and motor disturbances seen in rabbits exposed to cocaine in utero. Moreover, the long-term cognitive and motor deficits observed in the rabbit model are in agreement with the recent reports indicating that persistent attentional and other behavioral deficits may be evident in cocaine-exposed children as they grow older and are challenged to master more complex cognitive tasks.
Collapse
Affiliation(s)
- J A Harvey
- Department of Pharmacology and Physiology, MCP Hahnemann University, 3200 Henry Avenue, Philadelphia, PA 19129, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Frost DO, Cadet JL. Effects of methamphetamine-induced neurotoxicity on the development of neural circuitry: a hypothesis. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2000; 34:103-18. [PMID: 11113502 DOI: 10.1016/s0165-0173(00)00042-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Exposure of the developing brain to methamphetamine has well-studied biochemical and behavioral consequences. We review: (1) the effects of methamphetamine on mature serotonergic and dopaminergic pathways; (2) the mechanisms of methamphetamine neurotoxicity and (3) the role of serotonergic and dopaminergic signaling in sculpting developing neural circuitry. Consideration of these data suggest the types of neural circuit alterations that may result from exposure of the developing brain to methamphetamine and that may underlie functional defects.
Collapse
Affiliation(s)
- D O Frost
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, 655 West Baltimore St., Baltimore, MD 21201, USA.
| | | |
Collapse
|
32
|
Nicola SM, Surmeier J, Malenka RC. Dopaminergic modulation of neuronal excitability in the striatum and nucleus accumbens. Annu Rev Neurosci 2000; 23:185-215. [PMID: 10845063 DOI: 10.1146/annurev.neuro.23.1.185] [Citation(s) in RCA: 665] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The striatum and its ventral extension, the nucleus accumbens, are involved in behaviors as diverse as motor planning, drug seeking, and learning. Invariably, these striatally mediated behaviors depend on intact dopaminergic innervation. However, the mechanisms by which dopamine modulates neuronal function in the striatum and nucleus accumbens have been difficult to elucidate. Recent electrophysiological studies have revealed that dopamine alters both voltage-dependent conductances and synaptic transmission, resulting in state-dependent modulation of target cells. These studies make clear predictions about how dopamine, particularly via D1 receptor activation, should alter the responsiveness of striatal neurons to extrinsic excitatory synaptic activity.
Collapse
Affiliation(s)
- S M Nicola
- Department of Neurology, University of California at San Francisco 94143, USA.
| | | | | |
Collapse
|
33
|
In utero cocaine-induced dysfunction of dopamine D1 receptor signaling and abnormal differentiation of cerebral cortical neurons. J Neurosci 2000. [PMID: 10844030 DOI: 10.1523/jneurosci.20-12-04606.2000] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Monoamines modulate neuronal differentiation, and alteration of monoamine neurotransmission during development produces specific changes in neuronal structure, function, and pattern formation. We have previously observed that prenatal exposure to cocaine in a clinically relevant animal model produces increased length of pyramidal neuron dendrites in the anterior cingulate cortex (ACC) postnatally. We now report that cocaine administered intravenously to pregnant rabbits at gestational stages preceding and during cortical histogenesis results in the early onset of hypertrophic dendritic outgrowth in the embryonic ACC. Confocal microscopy of DiI-labeled neurons revealed that the atypical, tortuous dendritic profiles seen postnatally in ACC-cocaine neurons already are apparent in utero. No defects in neuronal growth were observed in visual cortex (VC), a region lacking prominent dopamine innervation. In striking correlation with our in vivo results, in vitro experiments revealed a significant enhancement of spontaneous process outgrowth of ACC neurons isolated from cocaine-exposed fetuses but no changes in neurons derived from visual cortex. The onset of modified growth in vivo is paralleled by reduced D(1A) receptor coupling to its G-protein. These data suggest that the dynamic growth of neurons can be regulated by early neurotransmitter signaling in a selective fashion. Prenatal onset of defects in dopamine receptor signaling contributes to abnormal circuit formation and may underlie specific cognitive and behavioral dysfunction.
Collapse
|
34
|
Zachor DA, Moore JF, Brezausek C, Theibert A, Percy AK. Cocaine inhibits NGF-induced PC12 cells differentiation through D(1)-type dopamine receptors. Brain Res 2000; 869:85-97. [PMID: 10865062 DOI: 10.1016/s0006-8993(00)02355-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In utero cocaine exposure can adversely affect CNS development. Previous studies showed that cocaine inhibits neuronal differentiation in a dose-dependent fashion in nerve growth factor (NGF)-stimulated PC12 cells. Cocaine binds with high affinity to several neurotransmitter transporters, resulting in elevated neurotransmitter levels in nerve endings. To determine if cocaine inhibits neurite outgrowth through the effects of these neurotransmitters, we applied dopamine, norepinephrine, serotonin, and acetylcholine to NGF-induced PC12 cells. Dopamine was the only neurotransmitter to inhibit neurite outgrowth significantly in a dose-dependent pattern without affecting cell viability. Norepinephrine and acetylcholine did not affect neurite outgrowth, while serotonin enhanced it. Furthermore, GBR 12909, a potent dopamine transporter (DAT) inhibitor, yielded similar effects. We then showed PC12 cells express D(1) and D(2) receptors and DAT proteins. Dopamine uptake measured over time was significantly blocked by cocaine and GBR 12909 which may result in elevated extracellular dopamine. The role of dopamine receptors in PC12 differentiation was further examined by using D(1) and D(2) specific receptor agonists. Only the D(1) agonist, SKF-38393, had a significant dose-dependent inhibitory effect. In addition, a D(1) antagonist produced significant recovery of neurite outgrowth in cocaine-treated cells. These findings suggest that cocaine inhibitory effects on neuronal differentiation are mediated through its binding to the dopamine transporter, resulting in increased dopamine level in the synapses. Subsequently, up regulation of D(1) receptors alters NGF signaling pathways.
Collapse
Affiliation(s)
- D A Zachor
- Department of Pediatrics, University of Alabama at Birmingham, 331C, Sparks Building, 1720 Seventh Avenue South, Birmingham, AL 35294-0017, USA.
| | | | | | | | | |
Collapse
|
35
|
Jones LB, Stanwood GD, Reinoso BS, Washington RA, Wang HY, Friedman E, Levitt P. In utero cocaine-induced dysfunction of dopamine D1 receptor signaling and abnormal differentiation of cerebral cortical neurons. J Neurosci 2000; 20:4606-14. [PMID: 10844030 PMCID: PMC6772445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Monoamines modulate neuronal differentiation, and alteration of monoamine neurotransmission during development produces specific changes in neuronal structure, function, and pattern formation. We have previously observed that prenatal exposure to cocaine in a clinically relevant animal model produces increased length of pyramidal neuron dendrites in the anterior cingulate cortex (ACC) postnatally. We now report that cocaine administered intravenously to pregnant rabbits at gestational stages preceding and during cortical histogenesis results in the early onset of hypertrophic dendritic outgrowth in the embryonic ACC. Confocal microscopy of DiI-labeled neurons revealed that the atypical, tortuous dendritic profiles seen postnatally in ACC-cocaine neurons already are apparent in utero. No defects in neuronal growth were observed in visual cortex (VC), a region lacking prominent dopamine innervation. In striking correlation with our in vivo results, in vitro experiments revealed a significant enhancement of spontaneous process outgrowth of ACC neurons isolated from cocaine-exposed fetuses but no changes in neurons derived from visual cortex. The onset of modified growth in vivo is paralleled by reduced D(1A) receptor coupling to its G-protein. These data suggest that the dynamic growth of neurons can be regulated by early neurotransmitter signaling in a selective fashion. Prenatal onset of defects in dopamine receptor signaling contributes to abnormal circuit formation and may underlie specific cognitive and behavioral dysfunction.
Collapse
Affiliation(s)
- L B Jones
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Ghil SH, Kim BJ, Lee YD, Suh-Kim H. Neurite outgrowth induced by cyclic AMP can be modulated by the alpha subunit of Go. J Neurochem 2000; 74:151-8. [PMID: 10617116 DOI: 10.1046/j.1471-4159.2000.0740151.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although abundant Go has been found in nervous tissues and it has been implicated in neuronal differentiation, the mechanism of how Go modulates neuronal differentiation has not been defined. Here, we report that the alpha subunit of Go (alphao) modulates neurite outgrowth by interfering with the signaling pathway initiated by cyclic AMP (cAMP). In F11 cells, cAMP induced neurite outgrowth and activated cAMP-responsive element binding protein (CREB). Specific inhibition of cAMP-dependent protein kinase reduced both CREB activity and neurite outgrowth (NOG). Interestingly, cAMP reduced phosphorylation of extracellular signal-regulated kinase (Erk). Neither a dominant negative form nor an active form of Ras altered neurite outgrowth. Expression of alphao (alphao(wt)) decreased the average length of neurites but increased the number of neurites per cell. An active mutant, alphaoQ205L, which lost GTPase activity and thus could not bind to Gbetagamma, gave similar results, suggesting that the effect of alphao is not mediated through Gbetagamma. Expression of ao(wt) or alphaoQ205L also prohibited CREB activation. Thus, activation of Erk may not be essential for neuronal differentiation in F11 cells and alphao may cause changes in NOG by inhibiting CREB activation.
Collapse
Affiliation(s)
- S H Ghil
- Department of Anatomy, School of Medicine, Ajou University, Suwon, South Korea
| | | | | | | |
Collapse
|
37
|
Abstract
Dopamine, an important neuromodulator in the retina, controls the balance of rod cone photoreceptor activity and influences the activity of several interneurons. The postnatal development of dopaminergic neurons, visualized immunocytochemically, was compared to the development of dopamine D1 receptor immunoreactivity. Expression of D1 receptors was monitored throughout the postnatal development of the rat retina using a subtype-specific monoclonal antibody. D1 receptors are expressed in the inner plexiform layer beginning at birth. Labeling of the inner plexiform layer changed from a diffuse pattern, staining the entire layer, to the typical adult punctate staining, that was organized in layered bands and occurred in the second postnatal week. The staining did not co-localize with dopaminergic cells; instead, it colocalized with cells in the inner nuclear layer or the ganglion cell layer. Within these cells, D1 receptors were most heavily expressed in processes stratifying in the inner plexiform layer. Staining in the outer plexiform layer and in horizontal cells was found beginning in the second postnatal week. Clustering of the D1 receptor within plexiform layers, a process typical for the well-described function of dopamine modulation in the adult, occurred late in postnatal development. A possible function of D1 receptors in neuronal development is discussed.
Collapse
Affiliation(s)
- P Koulen
- Max-Planck-Institut für Hirnforschung, Abteilung für Neuroanatomie, Frankfurt am Main, Germany.
| |
Collapse
|
38
|
Chergui K, Lacey MG. Modulation by dopamine D1-like receptors of synaptic transmission and NMDA receptors in rat nucleus accumbens is attenuated by the protein kinase C inhibitor Ro 32-0432. Neuropharmacology 1999; 38:223-31. [PMID: 10218863 DOI: 10.1016/s0028-3908(98)00187-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Dopamine, acting at a D1-like receptor, depresses the release of glutamate in the nucleus accumbens (NAcc) in brain slices, thereby reducing the amplitude of the excitatory postsynaptic current (EPSC). This effect depends upon an inhibitory feedback action of adenosine, liberated following facilitation of postsynaptic NMDA receptors by D1 receptor activation, an action independent of adenylyl cyclase stimulation or cyclic AMP-dependent protein kinase (PKA; Harvey, J., Lacey, M.G., 1997. J. Neurosci. 17, 5271). Using whole-cell recording from NAcc neurones, the dopamine depression of the EPSC was blocked by pre-treatment of brain slices with the selective protein kinase C (PKC) inhibitor Ro 32-0432, but only minimally attenuated by intracellular dialysis of single cells with Ro 32-0432 in the recording pipette. With synaptic transmission blocked by tetrodotoxin, inward currents caused by application of NMDA were enhanced by the D1 receptor agonist SKF 81297A in half the cells tested. In a separate population of cells dialysed intracellularly with Ro 32-0432, SKF 81297A was without effect on NMDA current amplitude. These findings indicate a functional role for phospholipase C-coupled D1-like receptors in both modulating synaptic transmission in NAcc and potentiating NMDA receptors on a subset of NAcc neurones, via PKC activation.
Collapse
Affiliation(s)
- K Chergui
- Department of Pharmacology, The Medical School, University of Birmingham, Edgbaston, UK
| | | |
Collapse
|
39
|
Pfeiffer-Linn CL, Lasater EM. Multiple second-messenger system modulation of voltage-activated calcium currents in teleost retinal horizontal cells. J Neurophysiol 1998; 80:377-88. [PMID: 9658058 DOI: 10.1152/jn.1998.80.1.377] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Two voltage-activated calcium currents, a transient T-type and a PL-sustained type, have been measured in isolated, cultured white bass horizontal cells. These two voltage-activated calcium currents were found to be modulated by two independent second-messenger systems. Furthermore, activation of either second-messenger system led to similar changes in calcium current activity. Activation of the cyclic AMP second-messenger pathway or the sn-1,2-diacylglycerol (DAG) second-messenger system resulted in a significant decrease in the amplitude of the transient current and a simultaneous large increase in the amplitude of the sustained current. Both second-messenger systems achieved their effects through protein phosphorylation. The cyclic AMP pathway resulted in the activation of protein kinase A (PKA) and the DAG pathway worked to activate protein kinase C (PKC). Two protein kinase inhibitors were analyzed in this study for their ability to inhibit second-messenger activated protein kinase activity and separate the two pathways. The peptide cyclic AMP-dependent protein kinase inhibitor and staurosporine were found to be nonspecific at high concentrations and inhibited both second-messenger pathways. At low concentrations however, staurosporine specifically inhibited only PKC, whereas adenosine 3',5'-cyclic monophosphate (cAMP)-dependent protein kinase inhibitor was selective for PKA. Both second-messenger systems were activated by the neuromodulator, dopamine. Thus one agonist can initiate multiple second-messenger systems leading to similar changes in voltage-activated calcium current activity. The modulatory action on calcium currents produced by one second-messenger system added to the modulatory action resulting from activation of the other second-messenger system. The effect is to alter the magnitude of the horizontal cell calcium currents.
Collapse
Affiliation(s)
- C L Pfeiffer-Linn
- John Moran Eye Center, University of Utah Health Sciences Center, Salt Lake City, Utah 84132, USA
| | | |
Collapse
|
40
|
Schmidt U, Pilgrim C, Beyer C. Differentiative effects of dopamine on striatal neurons involve stimulation of the cAMP/PKA pathway. Mol Cell Neurosci 1998; 11:9-18. [PMID: 9608529 DOI: 10.1006/mcne.1998.0668] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The neurotransmitter dopamine (DA) stimulates neurite outgrowth and growth cone formation in cultures of embryonic rat striatum through activation of D1 but not D2 receptors. We show here that neurite outgrowth could be stimulated to a similar extent by elevating cellular cAMP levels. Second, the neuritotrophic effect of DA was completely abolished by inhibiting adenylate cyclase or protein kinase A (PKA) but not protein kinase C (PKC). Third, double staining of cultures with antibodies against growth-associated protein-43 (GAP-43) and the phosphorylated form of the cAMP response element binding protein (pCREB) showed that pCREB was nearly exclusively associated with GAP-43-positive, i.e., actively growing, neurons. Again, this effect depended on D1 receptor and PKA activation. Although cross-talk with other signaling pathways needs to be studied further, we conclude that DA promotes the differentiation of striatal neurons via stimulation of D1 receptors and the cAMP/PKA signal transduction pathway.
Collapse
Affiliation(s)
- U Schmidt
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Germany
| | | | | |
Collapse
|
41
|
Missale C, Nash SR, Robinson SW, Jaber M, Caron MG. Dopamine receptors: from structure to function. Physiol Rev 1998; 78:189-225. [PMID: 9457173 DOI: 10.1152/physrev.1998.78.1.189] [Citation(s) in RCA: 2471] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The diverse physiological actions of dopamine are mediated by at least five distinct G protein-coupled receptor subtypes. Two D1-like receptor subtypes (D1 and D5) couple to the G protein Gs and activate adenylyl cyclase. The other receptor subtypes belong to the D2-like subfamily (D2, D3, and D4) and are prototypic of G protein-coupled receptors that inhibit adenylyl cyclase and activate K+ channels. The genes for the D1 and D5 receptors are intronless, but pseudogenes of the D5 exist. The D2 and D3 receptors vary in certain tissues and species as a result of alternative splicing, and the human D4 receptor gene exhibits extensive polymorphic variation. In the central nervous system, dopamine receptors are widely expressed because they are involved in the control of locomotion, cognition, emotion, and affect as well as neuroendocrine secretion. In the periphery, dopamine receptors are present more prominently in kidney, vasculature, and pituitary, where they affect mainly sodium homeostasis, vascular tone, and hormone secretion. Numerous genetic linkage analysis studies have failed so far to reveal unequivocal evidence for the involvement of one of these receptors in the etiology of various central nervous system disorders. However, targeted deletion of several of these dopamine receptor genes in mice should provide valuable information about their physiological functions.
Collapse
Affiliation(s)
- C Missale
- Department of Cell Biology, Howard Hughes Medical Institute Laboratories, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | |
Collapse
|
42
|
Shearman LP, Zeitzer J, Weaver DR. Widespread expression of functional D1-dopamine receptors in fetal rat brain. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1997; 102:105-15. [PMID: 9298239 DOI: 10.1016/s0165-3806(97)00091-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Maternal treatment with cocaine or the D1-dopamine receptor agonist, SKF 38393, induces expression of the immediate-early gene, c-fos, in fetal rodent brain. Our previous studies have focused on the suprachiasmatic nucleus late in gestation. In the present report, we examined the anatomical distribution of functional D1-dopamine receptors throughout fetal rat brain. Functional D1 receptors were defined using three complementary methods: in situ hybridization to detect D1 receptor mRNA, autoradiographic detection of 125I-SCH 23982 binding, and in situ hybridization to detect c-fos gene expression induced by maternal treatment with SKF 38393. D1-dopamine receptor binding, receptor mRNA, and SKF 38393-induced c-fos gene expression are widespread in fetal brain by late gestation. These data indicate that the fetal brain is sensitive to dopamine receptor activation, and suggest that gestational exposure to drugs of abuse acting via dopaminergic mechanisms may influence fetal brain function.
Collapse
Affiliation(s)
- L P Shearman
- Laboratory of Developmental Chronobiology, Massachusetts General Hospital, Jackson, Boston 02114, USA
| | | | | |
Collapse
|
43
|
A postsynaptic interaction between dopamine D1 and NMDA receptors promotes presynaptic inhibition in the rat nucleus accumbens via adenosine release. J Neurosci 1997. [PMID: 9204911 DOI: 10.1523/jneurosci.17-14-05271.1997] [Citation(s) in RCA: 140] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The mechanism underlying dopamine D1 receptor-mediated attenuation of glutamatergic synaptic input to nucleus accumbens (NAcc) neurons was investigated in slices of rat forebrain, using whole-cell patch-clamp recording. The depression by dopamine of EPSCs evoked by single-shock cortical stimulation was stimulus-dependent. Synaptic activation of NMDA-type glutamate receptors was critical for this effect, because dopamine-induced EPSC depressions were blocked by the competitive NMDA receptor antagonist D/L-2-amino-5-phosphonopentanoate (AP5). Application of NMDA also depressed the EPSC, and both this effect and the dopamine depressions were blocked by the A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), implicating adenosine release in the EPSC depression. A1 receptor agonists also depressed EPSCs by a presynaptic action, causing increased paired-pulse facilitation, but this was insensitive to AP5. Activation of D1 receptors enhanced both postsynaptic inward currents evoked by NMDA application and the isolated NMDA receptor-mediated component of synaptic transmission. The biochemical processes underlying the dopamine-induced EPSC depression did not involve either protein kinase A or the production of cAMP and its metabolites, because this effect was resistant to the protein kinase inhibitors H89 and H7 and the cAMP-specific phosphodiesterase inhibitor rolipram. We conclude that activation of postsynaptic D1 receptors enhances the synaptic activation of NMDA receptors in nucleus accumbens neurons, thereby promoting a transsynaptic feedback inhibition of glutamatergic synaptic transmission via release of adenosine. Unusually for D1 receptors, this phenomenon occurs independently of adenylyl cyclase stimulation. This process may contribute to the locomotor stimulant action of dopaminergic agents in the NAcc.
Collapse
|
44
|
Harvey J, Lacey MG. A postsynaptic interaction between dopamine D1 and NMDA receptors promotes presynaptic inhibition in the rat nucleus accumbens via adenosine release. J Neurosci 1997; 17:5271-80. [PMID: 9204911 PMCID: PMC6793812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The mechanism underlying dopamine D1 receptor-mediated attenuation of glutamatergic synaptic input to nucleus accumbens (NAcc) neurons was investigated in slices of rat forebrain, using whole-cell patch-clamp recording. The depression by dopamine of EPSCs evoked by single-shock cortical stimulation was stimulus-dependent. Synaptic activation of NMDA-type glutamate receptors was critical for this effect, because dopamine-induced EPSC depressions were blocked by the competitive NMDA receptor antagonist D/L-2-amino-5-phosphonopentanoate (AP5). Application of NMDA also depressed the EPSC, and both this effect and the dopamine depressions were blocked by the A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), implicating adenosine release in the EPSC depression. A1 receptor agonists also depressed EPSCs by a presynaptic action, causing increased paired-pulse facilitation, but this was insensitive to AP5. Activation of D1 receptors enhanced both postsynaptic inward currents evoked by NMDA application and the isolated NMDA receptor-mediated component of synaptic transmission. The biochemical processes underlying the dopamine-induced EPSC depression did not involve either protein kinase A or the production of cAMP and its metabolites, because this effect was resistant to the protein kinase inhibitors H89 and H7 and the cAMP-specific phosphodiesterase inhibitor rolipram. We conclude that activation of postsynaptic D1 receptors enhances the synaptic activation of NMDA receptors in nucleus accumbens neurons, thereby promoting a transsynaptic feedback inhibition of glutamatergic synaptic transmission via release of adenosine. Unusually for D1 receptors, this phenomenon occurs independently of adenylyl cyclase stimulation. This process may contribute to the locomotor stimulant action of dopaminergic agents in the NAcc.
Collapse
Affiliation(s)
- J Harvey
- Department of Pharmacology, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | | |
Collapse
|
45
|
Maclennan AJ, Marks L, Gaskin AA, Lee N. Embryonic expression pattern of H218, a G-protein coupled receptor homolog, suggests roles in early mammalian nervous system development. Neuroscience 1997; 79:217-24. [PMID: 9178877 DOI: 10.1016/s0306-4522(96)00601-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Heterologous expression studies employing mammalian cell tissue culture techniques and in vivo studies of lower eukaryotes suggest that G-protein coupled receptors may play critical roles in regulating early stages of vertebrate nervous system development. Previous work suggests that H218, a rat G-protein coupled receptor homolog, could serve such a role. Most importantly, northern blot data indicate that whole brain H218 mRNA levels are highest during embryogenesis. In the present studies we raised, affinity-purified and characterized several anti-H218, polyclonal antisera and immunohistochemically mapped the expression of H218 during the early stages of rat embryonic nervous system development. The resulting data indicate that H218 is preferentially expressed in young, differentiating neuronal cell bodies and axons. Moreover, the expression is temporally regulated such that highest H218 levels are found in neuronal cell bodies during their early stages of differentiation and in axons during their outgrowth. Therefore, we propose that H218 signal transduction may widely participate in the regulation of some of the first steps in neuronal differentiation including axon outgrowth.
Collapse
Affiliation(s)
- A J Maclennan
- Department of Neuroscience, University of Florida Brain Institute, University of Florida College of Medicine, Gainesville 32610-0244, U.S.A
| | | | | | | |
Collapse
|
46
|
Levitt P, Harvey JA, Friedman E, Simansky K, Murphy EH. New evidence for neurotransmitter influences on brain development. Trends Neurosci 1997; 20:269-74. [PMID: 9185309 DOI: 10.1016/s0166-2236(96)01028-4] [Citation(s) in RCA: 256] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The early appearance of monoamine systems in the developing mammalian CNS suggests that they play a role in neural development. We review data from two model systems that provide compelling new evidence of this role. In one model system-in utero exposure to cocaine-specific and robust alterations are seen in dopamine-rich areas of the cerebral cortex, such as the anterior cingulate cortex: D1 receptor-G protein coupling is greatly reduced, the GABAergic system is altered and pyramidal dendrites undergo excessive growth. In a second model system-a transgenic mouse line in which the gene that encodes monoamine oxidase A (MAOA) is disrupted, resulting in excessively high 5-HT levels-barrels fail to form in the developing somatosensory cortex. Both models reveal the effects of very early manipulation of monoamines on forebrain development, and the long-term anomalies that persist into adulthood.
Collapse
Affiliation(s)
- P Levitt
- Dept of Neurobiology, University of Pittsburgh School of Medicine, PA 15261, USA
| | | | | | | | | |
Collapse
|
47
|
Shelke RR, Lakshmana MK, Ramamohan Y, Raju TR. Levels of dopamine and noradrenaline in the developing of retina--effect of light deprivation. Int J Dev Neurosci 1997; 15:139-43. [PMID: 9099624 DOI: 10.1016/s0736-5748(96)00080-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The effect of light deprivation on the levels of dopamine and noradrenaline was studied in the developing rat retina. These transmitters were estimated in three groups of rats: (i) cycling light reared; (ii) dark reared since birth; and (iii) dark reared since birth, but exposed to cycling light for 1 day prior to the estimation of catecholamines. Our results show that (1) there is a progressive decrease in the levels of dopamine and noradrenaline in the cycling light and dark reared rats during postnatal development; (2) dark rearing further reduces the content of dopamine and noradrenaline; and (3) restoration of physiological (light) stimulus in the dark-reared rats during the early postnatal period results in the recovery of noradrenaline to a greater extent than that of dopamine. This study demonstrates a progressive decrease in the plasticity of dopaminergic system during retinal development, while such a decrease is not apparent in the noradrenergic system.
Collapse
Affiliation(s)
- R R Shelke
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, India
| | | | | | | |
Collapse
|
48
|
Kwon JH, Eves EM, Farrell S, Segovia J, Tobin AJ, Wainer BH, Downen M. Beta-adrenergic receptor activation promotes process outgrowth in an embryonic rat basal forebrain cell line and in primary neurons. Eur J Neurosci 1996; 8:2042-55. [PMID: 8921294 DOI: 10.1111/j.1460-9568.1996.tb00724.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A clonal cell line, AS583-8.E4.22, from the embryonic day 15 rat basal forebrain was established using retrovirus-mediated transduction of a temperature-sensitive mutant of the simian virus 40 (SV40) large tumour antigen. The cell line expresses cytoskeletal and neurotransmitter features indicative of neuronal commitment. In response to agents that increase intracellular cAMP, including forskolin and catecholamines, the cell line exhibits rapid process outgrowth and growth cone formation that does not require new gene expression or protein synthesis. The neurite outgrowth induced by catecholamines is mediated by beta 2-adrenergic receptors and is characterized by a rapid, reversible redistribution of filamentous actin. Neurons from primary cultures of embryonic day 15 basal forebrain were also found to respond to beta-adrenergic receptor agonists by enhancing growth cone formation. These results suggest that catecholamines provide cues that induce cytoskeletal rearrangements leading to neuronal process outgrowth and growth cone formation in the developing basal forebrain and possibly other neuronal progenitor cell populations. The neuronal basal forebrain cell line provides an ideal model to study the signalling mechanisms underlying the catecholamine-induced process outgrowth.
Collapse
Affiliation(s)
- J H Kwon
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Schmidt U, Beyer C, Oestreicher AB, Reisert I, Schilling K, Pilgrim C. Activation of dopaminergic D1 receptors promotes morphogenesis of developing striatal neurons. Neuroscience 1996; 74:453-60. [PMID: 8865196 DOI: 10.1016/0306-4522(96)00201-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The early dopaminergic input from the midbrain may play an important role in the development of the basal ganglia. We therefore investigated whether and how dopamine affects the morphogenesis of striatal target neurons. Dissociated cell cultures of embryonic day 17 rat striatum were raised for seven days. Cells were then incubated with dopamine or various receptor-specific ligands for 1 h. At various times after termination of the treatment, cells were immunostained for growth-associated protein-43. Morphological parameters including numbers of growth cones, length of neurites, number of bifurcations, and neuronal soma size were assessed by means of a computer-based morphometric device. Treatment with dopamine in low concentrations as well as with the D1-like receptor agonist SKF 38393 increased the numbers of growth cones and neurite length and arborization. The morphogenetic effect took several hours to evolve and remained stable for at least 24 h. It could be blocked by the D1-like receptor antagonist SCH 23390 or by cycloheximide but not by pretreatment of the cultures with tetrodotoxin. The D2-like receptor agonist quinpirole had no effect on the morphological parameters and did not contribute to that of SKF 38393. Dopamine and SKF 38393 but not quinpirole also induced an increase in the number of neurons immunoreactive for Fos-like proteins. However, this effect was restricted to growth-associated protein-43-negative neurons. This is the first observation of a positive regulatory effect of D1-like receptors on neuronal morphogenesis. We conclude that the changes reflect true differentiation rather than short-term modulation of cellular properties and that c-fos induction is not an obligatory step in the transduction pathway coupling D1-like receptors to neurite outgrowth. Our results suggest that the differentiation of embryonic striatal neurons is promoted by the dopaminergic nigrostriatal projection through D1-like receptors.
Collapse
Affiliation(s)
- U Schmidt
- Abteilung Anatomie und Zellbiologie, Universität Ulm, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Yu PY, Eisner GM, Yamaguchi I, Mouradian MM, Felder RA, Jose PA. Dopamine D1A receptor regulation of phospholipase C isoform. J Biol Chem 1996; 271:19503-8. [PMID: 8702641 DOI: 10.1074/jbc.271.32.19503] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In LTK- cells stably transfected with rat D1A receptor cDNA, fenoldopam, a D1 agonist, increased phosphatidylinositol 4, 5-bisphosphate hydrolysis in a time-dependent manner. In the cytosol, phospholipase C (PLC) activity increased (50 +/- 7%) in 30 s, returned to basal level at 4 h, and decreased below basal values by 24 h; in the membrane, PLC activity also increased (36 +/- 13%) in 30 s, returned to basal level at 10 min, and decreased below basal value at 4 and 24 h. Fenoldopam also increased PLC-gamma protein in a time-dependent manner. The latter was blocked by the D1 antagonist SKF83742 and by a D1A antisense oligodeoxynucleotide, indicating involvement of the D1A receptor. The fenoldopam-induced increase in PLC-gamma and activity was mediated by protein kinase A (PKA) since it was blocked by the PKA antagonist Rp-8-CTP-adenosine cyclic 3':5'-monophosphorothioate (Rp-8-CTP-cAMP-S) and mimicked by direct stimulation of adenylyl cyclase with forskolin or by a PKA agonist, Sp-cAMP-S. Protein kinase C (PKC) was also involved, since the fenoldopam-induced increase in PLC-gamma protein was blocked by two different PKC inhibitors, calphostin C and chelerythrine; calphostin C also blocked the fenoldopam-induced increase in PLC activity. In addition, forskolin and a PKA agonist, Sp-8-CTP-cAMP-S, increased PKC activity, and direct stimulation of PKC with phorbol 12-myristate 13-acetate increased PLC-gamma protein and activity, effects that were blocked by calphostin C. We suggest that the D1A-mediated stimulation of PLC occurs as a result of PKA activation. PKA then stimulates PLC-gamma in cytosol and membrane via activation of PKC.
Collapse
Affiliation(s)
- P Y Yu
- Department of Pediatrics, Georgetown University Medical Center, Washington, D. C. 20007, USA
| | | | | | | | | | | |
Collapse
|