1
|
Laroche S, Stil A, Germain P, Cherif H, Chemtob S, Bouchard JF. Participation of L-Lactate and Its Receptor HCAR1/GPR81 in Neurovisual Development. Cells 2021; 10:1640. [PMID: 34208876 PMCID: PMC8303161 DOI: 10.3390/cells10071640] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 11/16/2022] Open
Abstract
During the development of the retina and the nervous system, high levels of energy are required by the axons of retinal ganglion cells (RGCs) to grow towards their brain targets. This energy demand leads to an increase of glycolysis and L-lactate concentrations in the retina. L-lactate is known to be the endogenous ligand of the GPR81 receptor. However, the role of L-lactate and its receptor in the development of the nervous system has not been studied in depth. In the present study, we used immunohistochemistry to show that GPR81 is localized in different retinal layers during development, but is predominantly expressed in the RGC of the adult rodent. Treatment of retinal explants with L-lactate or the exogenous GPR81 agonist 3,5-DHBA altered RGC growth cone (GC) morphology (increasing in size and number of filopodia) and promoted RGC axon growth. These GPR81-mediated modifications of GC morphology and axon growth were mediated by protein kinases A and C, but were absent in explants from gpr81-/- transgenic mice. Living gpr81-/- mice showed a decrease in ipsilateral projections of RGCs to the dorsal lateral geniculate nucleus (dLGN). In conclusion, present results suggest that L-lactate and its receptor GPR81 play an important role in the development of the visual nervous system.
Collapse
Affiliation(s)
- Samuel Laroche
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| | - Aurélie Stil
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| | - Philippe Germain
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| | - Hosni Cherif
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| | - Sylvain Chemtob
- Department of Pediatrics, Research Center-CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada;
- Department of Ophtalmology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Jean-François Bouchard
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| |
Collapse
|
2
|
Wu W, Wang Z, Xu K, Zhang X, Amei A, Gelernter J, Zhao H, Justice AC, Wang Z. Retrospective Association Analysis of Longitudinal Binary Traits Identifies Important Loci and Pathways in Cocaine Use. Genetics 2019; 213:1225-1236. [PMID: 31591132 PMCID: PMC6893384 DOI: 10.1534/genetics.119.302598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022] Open
Abstract
Longitudinal phenotypes have been increasingly available in genome-wide association studies (GWAS) and electronic health record-based studies for identification of genetic variants that influence complex traits over time. For longitudinal binary data, there remain significant challenges in gene mapping, including misspecification of the model for phenotype distribution due to ascertainment. Here, we propose L-BRAT (Longitudinal Binary-trait Retrospective Association Test), a retrospective, generalized estimating equation-based method for genetic association analysis of longitudinal binary outcomes. We also develop RGMMAT, a retrospective, generalized linear mixed model-based association test. Both tests are retrospective score approaches in which genotypes are treated as random conditional on phenotype and covariates. They allow both static and time-varying covariates to be included in the analysis. Through simulations, we illustrated that retrospective association tests are robust to ascertainment and other types of phenotype model misspecification, and gain power over previous association methods. We applied L-BRAT and RGMMAT to a genome-wide association analysis of repeated measures of cocaine use in a longitudinal cohort. Pathway analysis implicated association with opioid signaling and axonal guidance signaling pathways. Lastly, we replicated important pathways in an independent cocaine dependence case-control GWAS. Our results illustrate that L-BRAT is able to detect important loci and pathways in a genome scan and to provide insights into genetic architecture of cocaine use.
Collapse
Affiliation(s)
- Weimiao Wu
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut 06520
| | - Zhong Wang
- Baker Institute for Animal Health, Cornell University, Ithaca, New York 14850
| | - Ke Xu
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut 06511
- VA Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Xinyu Zhang
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut 06511
- VA Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Amei Amei
- Department of Mathematical Sciences, University of Nevada, Las Vegas, Nevada 89154
| | - Joel Gelernter
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut 06511
- VA Connecticut Healthcare System, West Haven, Connecticut 06516
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut 06520
| | - Amy C Justice
- VA Connecticut Healthcare System, West Haven, Connecticut 06516
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut 06511
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut 06520
| |
Collapse
|
3
|
Yuferov V, Zhang Y, Liang Y, Zhao C, Randesi M, Kreek MJ. Oxycodone Self-Administration Induces Alterations in Expression of Integrin, Semaphorin and Ephrin Genes in the Mouse Striatum. Front Psychiatry 2018; 9:257. [PMID: 29946272 PMCID: PMC6005861 DOI: 10.3389/fpsyt.2018.00257] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 05/24/2018] [Indexed: 11/28/2022] Open
Abstract
Oxycodone is one a commonly used medication for pain, and is also a widely abused prescription opioid, like other short-acting MOPr agonists. Neurochemical and structural adaptations in brain following chronic MOPr-agonist administration are thought to underlie pathogenesis and persistence of opiate addiction. Many axon guidance molecules, such as integrins, semaphorins, and ephrins may contribute to oxycodone-induced neuroadaptations through alterations in axon-target connections and synaptogenesis, that may be implicated in the behaviors associated with opiate addiction. However, little is known about this important area. The aim of this study is to investigate alterations in expression of selected integrin, semaphorin, ephrins, netrin, and slit genes in the nucleus accumbens (NAc) and caudate putamen (CPu) of mice following extended 14-day oxycodone self-administration (SA), using RNAseq. Methods: Total RNA from the NAc and CPu were isolated from adult male C57BL/6J mice within 1 h after the last session of oxycodone in a 14-day self-administration paradigm (4h/day, 0.25 mg/kg/infusion, FR1) or from yoked saline controls. Gene expressions were examined using RNA sequencing (RNA-Seq) technology. RNA-Seq libraries were prepared using Illumina's TruSeq® Stranded Total RNA LT kit. The reads were aligned to the mouse reference genome (version mm10) using STAR. DESeq2 was applied to the counts of protein coding genes to estimate the fold change between the treatment groups. False Discovery Rate (FDR) q < 0.1 were used to select genes that have a significant expression change. For selection of a subset of genes related to axon guidance pathway, REACTOME was used. Results: Among 38 known genes of the integrin, semaphorin, and ephrin gene families, RNA-seq data revealed up-regulation of six genes in the NAc: heterodimer receptor, integrins Itgal, Itgb2, and Itgam, and its ligand semaphorin Sema7a, two semaphorin receptors, plexins Plxnd1 and Plxdc1. There was down-regulation of eight genes in this region: two integrin genes Itga3 and Itgb8, semaphorins Sema3c, Sema4g, Sema6a, Sema6d, semaphorin receptor neuropilin Nrp2, and ephrin receptor Epha3. In the CPu, there were five differentially expressed axon guidance genes: up-regulation of three integrin genes, Itgal, Itgb2, Itga1, and down-regulation of Itga9 and ephrin Efna3 were thus observed. No significant alterations in expression of Netrin-1 or Slit were observed. Conclusion: We provide evidence for alterations in the expression of selective axon guidance genes in adult mouse brain following chronic self-administration of oxycodone. Further examination of oxycodone-induced changes in the expression of these specific axon guidance molecules and integrin genes in relation to behavior may provide new insights into development of addiction to oxycodone.
Collapse
Affiliation(s)
- Vadim Yuferov
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Yong Zhang
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Yupu Liang
- Research Bioinformatics, Clinical and Translational Science Award, The Rockefeller University, New York, NY, United States
| | - Connie Zhao
- Genomic Resource Center, The Rockefeller University, New York, NY, United States
| | - Matthew Randesi
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Mary J Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| |
Collapse
|
4
|
Wang J, Galvao J, Beach KM, Luo W, Urrutia RA, Goldberg JL, Otteson DC. Novel Roles and Mechanism for Krüppel-like Factor 16 (KLF16) Regulation of Neurite Outgrowth and Ephrin Receptor A5 (EphA5) Expression in Retinal Ganglion Cells. J Biol Chem 2016; 291:18084-95. [PMID: 27402841 DOI: 10.1074/jbc.m116.732339] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Indexed: 11/06/2022] Open
Abstract
Regenerative medicine holds great promise for the treatment of degenerative retinal disorders. Krüppel-like factors (KLFs) are transcription factors that have recently emerged as key tools in regenerative medicine because some of them can function as epigenetic reprogrammers in stem cell biology. Here, we show that KLF16, one of the least understood members of this family, is a POU4F2 independent transcription factor in retinal ganglion cells (RGCs) as early as embryonic day 15. When overexpressed, KLF16 inhibits RGC neurite outgrowth and enhances RGC growth cone collapse in response to exogenous ephrinA5 ligands. Ephrin/EPH signaling regulates RGC connectivity. The EphA5 promoter contains multiple GC- and GT-rich KLF-binding sites, which, as shown by ChIP-assays, bind KLF16 in vivo In electrophoretic mobility shift assays, KLF16 binds specifically to a single KLF site near the EphA5 transcription start site that is required for KLF16 transactivation. Interestingly, methylation of only six of 98 CpG dinucleotides within the EphA5 promoter blocks its transactivation by KLF16 but enables transactivation by KLF2 and KLF15. These data demonstrate a role for KLF16 in regulation of RGC neurite outgrowth and as a methylation-sensitive transcriptional regulator of EphA5 expression. Together, these data identify differential low level methylation as a novel mechanism for regulating KLF16-mediated EphA5 expression across the retina. Because of the critical role of ephrin/EPH signaling in patterning RGC connectivity, understanding the role of KLFs in regulating neurite outgrowth and Eph receptor expression will be vital for successful restoration of functional vision through optic nerve regenerative therapies.
Collapse
Affiliation(s)
- Jianbo Wang
- From the Departments of Physiological Optics and Vision Science and
| | - Joana Galvao
- the Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, California 94303, the Shiley Eye Institute, University of California San Diego, La Jolla, California 92093, and
| | - Krista M Beach
- From the Departments of Physiological Optics and Vision Science and
| | - Weijia Luo
- Biology and Biochemistry, University of Houston, Houston, Texas 77204
| | - Raul A Urrutia
- the Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Epigenomics Translational Program, Center for Individualized Medicine, Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905
| | - Jeffrey L Goldberg
- the Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, California 94303, the Shiley Eye Institute, University of California San Diego, La Jolla, California 92093, and
| | - Deborah C Otteson
- From the Departments of Physiological Optics and Vision Science and Biology and Biochemistry, University of Houston, Houston, Texas 77204,
| |
Collapse
|
5
|
A Serotonin Circuit Acts as an Environmental Sensor to Mediate Midline Axon Crossing through EphrinB2. J Neurosci 2016; 35:14794-808. [PMID: 26538650 DOI: 10.1523/jneurosci.1295-15.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Modulation of connectivity formation in the developing brain in response to external stimuli is poorly understood. Here, we show that the raphe nucleus and its serotonergic projections regulate pathfinding of commissural axons in zebrafish. We found that the raphe neurons extend projections toward midline-crossing axons and that when serotonergic signaling is blocked by pharmacological inhibition or by raphe neuron ablation, commissural pathfinding is disrupted. We demonstrate that the serotonin receptor htr2a is expressed on these commissural axons and that genetic knock-down of htr2a disrupts crossing. We further show that knock-down of htr2a or ablation of the raphe neurons increases ephrinB2a protein levels in commissural axons. An ephrinB2a mutant can rescue midline crossing when serotonergic signaling is blocked. Furthermore, we found that regulation of serotonin expression in the raphe neurons is modulated in response to the developmental environment. Hypoxia causes the raphe to decrease serotonin levels, leading to a reduction in midline crossing. Increasing serotonin in the setting of hypoxia restored midline crossing. Our findings demonstrate an instructive role for serotonin in axon guidance acting through ephrinB2a and reveal a novel mechanism for developmental interpretation of the environmental milieu in the generation of mature neural circuitry. SIGNIFICANCE STATEMENT We show here that serotonin has a novel role in regulating connectivity in response to the developmental environment. We demonstrate that serotonergic projections from raphe neurons regulate pathfinding of crossing axons. The neurons modulate their serotonin levels, and thus alter crossing, in response to the developmental environment including hypoxia. The findings suggest that modification of the serotonergic system by early exposures may contribute to permanent CNS connectivity alterations. This has important ramifications because of the association between premature birth and accompanying hypoxia, and increased risk of autism and evidence associating in utero exposure to some antidepressants and neurodevelopmental disorders. Finally, this work demonstrates that the vertebrate CNS can modulate its connectivity in response to the external environment.
Collapse
|
6
|
Dong Y. Silent Synapse-Based Circuitry Remodeling in Drug Addiction. Int J Neuropsychopharmacol 2015; 19:pyv136. [PMID: 26721952 PMCID: PMC4886671 DOI: 10.1093/ijnp/pyv136] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/14/2015] [Indexed: 12/01/2022] Open
Abstract
Exposure to cocaine, and likely other drugs of abuse, generates α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor-silent glutamatergic synapses in the nucleus accumbens. These immature synaptic contacts evolve after drug withdrawal to redefine the neurocircuital properties. These results raise at least three critical questions: (1) what are the molecular and cellular mechanisms that mediate drug-induced generation of silent synapses; (2) how are neurocircuits remodeled upon generation and evolution of drug-generated silent synapses; and (3) what behavioral consequences are produced by silent synapse-based circuitry remodeling? This short review analyzes related experimental results, and extends them to some speculations.
Collapse
Affiliation(s)
- Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pennsylvania (Dr Dong).
| |
Collapse
|
7
|
Abstract
In this issue of Science Signaling, Villanueva et al. report on the identification of heterotrimeric guanine nucleotide-binding protein (G protein) Gαi2 as an essential effector of Smoothened-mediated mammary epithelial cell proliferation. Through a series of in vivo experiments, the authors delineate a Smoothened-regulated Gli-independent Gαi signal that provides direct genetic evidence connecting Smoothened with Gαi in mice.
Collapse
Affiliation(s)
- Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
8
|
Gutekunst CA, Gross RE. Plexin a4 expression in adult rat cranial nerves. J Chem Neuroanat 2014; 61-62:13-9. [PMID: 24970554 PMCID: PMC4267999 DOI: 10.1016/j.jchemneu.2014.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/21/2014] [Accepted: 06/16/2014] [Indexed: 12/16/2022]
Abstract
PlexinsA1-A4 participate in class 3 semaphorin signaling as co-receptors to neuropilin 1 and 2. PlexinA4 is the latest member of the PlexinA subfamily to be identified. In previous studies, we described the expression of PlexinA4 in the brain and spinal cord of the adult rat. Here, antibodies to PlexinA4 were used to reveal immunolabeling in most of the cranial nerve surveyed. Labeling was found in the olfactory, optic, oculomotor, trochlear, trigeminal, abducens, facial, vestibulocochlear, glossopharyngeal, vagus, and hypoglossal nerves. This is the first detailed description of the cellular and subcellular distribution of PlexinA4 in the adult cranial nerves. The findings will set the basis for future studies on the potential role of PlexinA4 in regeneration and repair of the adult central and peripheral nervous system.
Collapse
Affiliation(s)
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
9
|
Dopamine challenge reveals neuroadaptive changes in marijuana abusers. Proc Natl Acad Sci U S A 2014; 111:11915-6. [PMID: 25114244 DOI: 10.1073/pnas.1412314111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
10
|
Xu H, Ferreira MM, Heilshorn SC. Small-molecule axon-polarization studies enabled by a shear-free microfluidic gradient generator. LAB ON A CHIP 2014; 14:2047-56. [PMID: 24781157 PMCID: PMC4528973 DOI: 10.1039/c4lc00162a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
A deep understanding of the mechanisms behind neurite polarization and axon path-finding is important for interpreting how the human body guides neurite growth during development and response to injury. Further, it is of great clinical importance to identify diffusible chemical cues that promote neurite regeneration for nervous tissue repair. Despite the fast development of various types of concentration gradient generators, it has been challenging to fabricate neuron-friendly (i.e. shear-free and biocompatible for neuron growth and maturation) devices to create stable gradients, particularly for fast diffusing small molecules, which typically require high flow and shear rates. Here we present a finite element analysis for a polydimethylsiloxane/polyethylene glycol diacrylate (PDMS/PEG-DA) based gradient generator, describe the microfabrication process, and validate its use for neuronal axon polarization studies. This device provides a totally shear-free, biocompatible microenvironment with a linear and stable concentration gradient of small molecules such as forskolin. The gradient profile in this device can be customized by changing the composition or width of the PEG-DA barriers during direct UV photo-patterning within a permanently bonded PDMS device. Primary rat cortical neurons (embryonic E18) exposed to soluble forskolin gradients for 72 h exhibited statistically significant polarization and guidance of their axons. This device provides a useful platform for both chemotaxis and directional guidance studies, particularly for shear sensitive and non-adhesive cell cultures, while allowing fast new device design prototyping at a low cost.
Collapse
Affiliation(s)
- Hui Xu
- Department of Materials Science and Engineering, Stanford Cardiovascular Institute, Stanford University, 476 Lomita Mall, McCullough Building, Stanford, CA 94305-4045, USA.
| | | | | |
Collapse
|
11
|
unfulfilled interacting genes display branch-specific roles in the development of mushroom body axons in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2014; 4:693-706. [PMID: 24558265 PMCID: PMC4577660 DOI: 10.1534/g3.113.009829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The mushroom body (MB) of Drosophila melanogaster is an organized collection of interneurons that is required for learning and memory. Each of the three subtypes of MB neurons, γ, α´/β´, and α/β, branch at some point during their development, providing an excellent model in which to study the genetic regulation of axon branching. Given the sequential birth order and the unique patterning of MB neurons, it is likely that specific gene cascades are required for the different guidance events that form the characteristic lobes of the MB. The nuclear receptor UNFULFILLED (UNF), a transcription factor, is required for the differentiation of all MB neurons. We have developed and used a classical genetic suppressor screen that takes advantage of the fact that ectopic expression of unf causes lethality to identify candidate genes that act downstream of UNF. We hypothesized that reducing the copy number of unf-interacting genes will suppress the unf-induced lethality. We have identified 19 candidate genes that when mutated suppress the unf-induced lethality. To test whether candidate genes impact MB development, we performed a secondary phenotypic screen in which the morphologies of the MBs in animals heterozygous for unf and a specific candidate gene were analyzed. Medial MB lobes were thin, missing, or misguided dorsally in five double heterozygote combinations (;unf/+;axin/+, unf/+;Fps85D/+, ;unf/+;Tsc1/+, ;unf/+;Rheb/+, ;unf/+;msn/+). Dorsal MB lobes were missing in ;unf/+;DopR2/+ or misprojecting beyond the termination point in ;unf/+;Sytβ double heterozygotes. These data suggest that unf and unf-interacting genes play specific roles in axon development in a branch-specific manner.
Collapse
|
12
|
Sidharthan NP, Minchin RF, Butcher NJ. Cytosolic sulfotransferase 1A3 is induced by dopamine and protects neuronal cells from dopamine toxicity: role of D1 receptor-N-methyl-D-aspartate receptor coupling. J Biol Chem 2013; 288:34364-74. [PMID: 24136195 DOI: 10.1074/jbc.m113.493239] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Dopamine neurotoxicity is associated with several neurodegenerative diseases, and neurons utilize several mechanisms, including uptake and metabolism, to protect them from injury. Metabolism of dopamine involves three enzymes: monoamine oxidase, catechol O-methyltransferase, and sulfotransferase. In primates but not lower order animals, a sulfotransferase (SULT1A3) is present that can rapidly metabolize dopamine to dopamine sulfate. Here, we show that SULT1A3 and a closely related protein SULT1A1 are highly inducible by dopamine. This involves activation of the D1 and NMDA receptors. Both ERK1/2 phosphorylation and calcineurin activation are required for induction. Pharmacological agents that inhibited induction or siRNA targeting SULT1A3 significantly increased the susceptibility of cells to dopamine toxicity. Taken together, these results show that dopamine can induce its own metabolism and protect neuron-like cells from damage, suggesting that SULT1A3 activity may be a risk factor for dopamine-dependent neurodegenerative diseases.
Collapse
Affiliation(s)
- Neelima P Sidharthan
- From the School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia 4072
| | | | | |
Collapse
|
13
|
Lind PA, Zhu G, Montgomery GW, Madden PAF, Heath AC, Martin NG, Slutske WS. Genome-wide association study of a quantitative disordered gambling trait. Addict Biol 2013; 18:511-22. [PMID: 22780124 DOI: 10.1111/j.1369-1600.2012.00463.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Disordered gambling is a moderately heritable trait, but the underlying genetic basis is largely unknown. We performed a genome-wide association study (GWAS) for disordered gambling using a quantitative factor score in 1312 twins from 894 Australian families. Association was conducted for 2 381 914 single-nucleotide polymorphisms (SNPs) using the family-based association test in Merlin followed by gene and pathway enrichment analyses. Although no SNP reached genome-wide significance, six achieved P-values < 1 × 10(-5) with variants in three genes (MT1X, ATXN1 and VLDLR) implicated in disordered gambling. Secondary case-control analyses found two SNPs on chromosome 9 (rs1106076 and rs12305135 near VLDLR) and rs10812227 near FZD10 on chromosome 12 to be significantly associated with lifetime Diagnostic and Statistical Manual of Mental Disorders, fourth edition pathological gambling and South Oaks Gambling Screen classified probable pathological gambling status. Furthermore, several addiction-related pathways were enriched for SNPs associated with disordered gambling. Finally, gene-based analysis of 24 candidate genes for dopamine agonist-induced gambling in individuals with Parkinson's disease suggested an enrichment of SNPs associated with disordered gambling. We report the first GWAS of disordered gambling. While further replication is required, the identification of susceptibility loci and biological pathways will be important in characterizing the biological mechanisms that underpin disordered gambling.
Collapse
Affiliation(s)
- Penelope A Lind
- Quantitative Genetics, Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | | | | | | | | | | | | |
Collapse
|
14
|
Prestoz L, Jaber M, Gaillard A. Dopaminergic axon guidance: which makes what? Front Cell Neurosci 2012; 6:32. [PMID: 22866028 PMCID: PMC3408579 DOI: 10.3389/fncel.2012.00032] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 07/15/2012] [Indexed: 01/30/2023] Open
Abstract
Mesotelencephalic pathways in the adult central nervous system have been studied in great detail because of their implication in major physiological functions as well as in psychiatric, neurological, and neurodegenerative diseases. However, the ontogeny of these pathways and the molecular mechanisms that guide dopaminergic axons during embryogenesis have been only recently studied. This line of research is of crucial interest for the repair of lesioned circuits in adulthood following neurodegenerative diseases or common traumatic injuries. For instance, in the adult, the anatomic and functional repair of the nigrostriatal pathway following dopaminergic embryonic neuron transplantation suggests that specific guidance cues exist which govern embryonic fibers outgrowth, and suggests that axons from transplanted embryonic cells are able to respond to theses cues, which then guide them to their final targets. In this review, we first synthesize the work that has been performed in the last few years on developing mesotelencephalic pathways, and summarize the current knowledge on the identity of cellular and molecular signals thought to be involved in establishing mesotelencephalic dopaminergic neuronal connectivity during embryogenesis in the central nervous system of rodents. Then, we review the modulation of expression of these molecular signals in the lesioned adult brain and discuss their potential role in remodeling the mesotelencephalic dopaminergic circuitry, with a particular focus on Parkinson's disease (PD). Identifying guidance molecules involved in the connection of grafted cells may be useful for cellular therapy in Parkinsonian patients, as these molecules may help direct axons from grafted cells along the long distance they have to travel from the substantia nigra to the striatum.
Collapse
Affiliation(s)
- Laetitia Prestoz
- Experimental and Clinical Neurosciences Laboratory, Research Group on Cellular Therapies in Brain Diseases, INSERM U1084, University of PoitiersPoitiers, France.
| | | | | |
Collapse
|
15
|
Johnstone AL, Reierson GW, Smith RP, Goldberg JL, Lemmon VP, Bixby JL. A chemical genetic approach identifies piperazine antipsychotics as promoters of CNS neurite growth on inhibitory substrates. Mol Cell Neurosci 2012; 50:125-35. [PMID: 22561309 DOI: 10.1016/j.mcn.2012.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 03/23/2012] [Accepted: 04/20/2012] [Indexed: 01/22/2023] Open
Abstract
Injury to the central nervous system (CNS) can result in lifelong loss of function due in part to the regenerative failure of CNS neurons. Inhibitory proteins derived from myelin and the astroglial scar are major barriers for the successful regeneration of injured CNS neurons. Previously, we described the identification of a novel compound, F05, which promotes neurite growth from neurons challenged with inhibitory substrates in vitro, and promotes axonal regeneration in vivo (Usher et al., 2010). To identify additional regeneration-promoting compounds, we used F05-induced gene expression profiles to query the Broad Institute Connectivity Map, a gene expression database of cells treated with >1300 compounds. Despite no shared chemical similarity, F05-induced changes in gene expression were remarkably similar to those seen with a group of piperazine phenothiazine antipsychotics (PhAPs). In contrast to antipsychotics of other structural classes, PhAPs promoted neurite growth of CNS neurons challenged with two different glial derived inhibitory substrates. Our pharmacological studies suggest a mechanism whereby PhAPs promote growth through antagonism of calmodulin signaling, independent of dopamine receptor antagonism. These findings shed light on mechanisms underlying neurite-inhibitory signaling, and suggest that clinically approved antipsychotic compounds may be repurposed for use in CNS injured patients.
Collapse
Affiliation(s)
- Andrea L Johnstone
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1400 NW 12th Ave, Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
16
|
Yen HK, Lin TF, Tseng IC. Detection and quantification of major toxigenic Microcystis genotypes in Moo-Tan reservoir and associated water treatment plant. ACTA ACUST UNITED AC 2012; 14:687-96. [DOI: 10.1039/c1em10389j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
17
|
Sillivan SE, Konradi C. Expression and function of dopamine receptors in the developing medial frontal cortex and striatum of the rat. Neuroscience 2011; 199:501-14. [PMID: 22015925 PMCID: PMC3253459 DOI: 10.1016/j.neuroscience.2011.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 10/03/2011] [Accepted: 10/04/2011] [Indexed: 12/16/2022]
Abstract
The timeline of dopamine (DA) system maturation and the signaling properties of DA receptors (DRs) during rat brain development are not fully characterized. We used in situ hybridization and quantitative PCR to map DR mRNA transcripts in the medial frontal cortex (mFC) and striatum (STR) of the rat from embryonic day (E) 15 to E21. The developmental trajectory of DR mRNAs revealed distinct patterns of DA receptors 1 and 2 (DRD1, DRD2) in these brain regions. Whereas the mFC had a steeper increase in DRD1 mRNA, the STR had a steeper increase in DRD2 mRNA. Both DR mRNAs were expressed at a higher level in the STR compared with the mFC. To identify the functional properties of DRs during embryonic development, the phosphorylation states of cyclic AMP response element binding protein, extracellular signal-regulated kinase 1/2, and glycogen synthase kinase 3 beta were examined after DR stimulation in primary neuronal cultures obtained from E15 and E18 embryos and cultured for 3 days to ensure a stable baseline level. DR-mediated signaling cascades were functional in E15 cultures in both brain regions. Because DA fibers do not reach the mFC by E15, and DA was not present in cultures, these data indicate that DRs can become functional in the absence of DA innervation. Because activation of DR signal transduction pathways can affect network organization of the developing brain, maternal exposure to drugs that affect DR activity may be liable to interfere with fetal brain development.
Collapse
Affiliation(s)
- Stephanie E. Sillivan
- Neuroscience Graduate Program, Vanderbilt University, Nashville, Tennessee, 37232, USA
| | - Christine Konradi
- Neuroscience Graduate Program, Vanderbilt University, Nashville, Tennessee, 37232, USA
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232, USA
- Center for Molecular Neuroscience, Vanderbilt University, Nashville, Tennessee, 37232, USA
- Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee, 37203, USA
| |
Collapse
|
18
|
Sillivan SE, Black YD, Naydenov AV, Vassoler FR, Hanlin RP, Konradi C. Binge cocaine administration in adolescent rats affects amygdalar gene expression patterns and alters anxiety-related behavior in adulthood. Biol Psychiatry 2011; 70:583-92. [PMID: 21571252 PMCID: PMC3159046 DOI: 10.1016/j.biopsych.2011.03.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 03/22/2011] [Accepted: 03/23/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND Administration of cocaine during adolescence alters neurotransmission and behavioral sensitization in adulthood, but the effect on the acquisition of fear memories and the development of emotion-based neuronal circuits is unknown. METHODS We examined fear learning and anxiety-related behaviors in adult male rats that were subjected to binge cocaine treatment during adolescence. We furthermore conducted gene expression analyses of the amygdala 22 hours after the last cocaine injection to identify molecular patterns that might lead to altered emotional processing. RESULTS Rats injected with cocaine during adolescence displayed less anxiety in adulthood than their vehicle-injected counterparts. In addition, cocaine-exposed animals were deficient in their ability to develop contextual fear responses. Cocaine administration caused transient gene expression changes in the Wnt signaling pathway, of axon guidance molecules, and of synaptic proteins, suggesting that cocaine perturbs dendritic structures and synapses in the amygdala. Phosphorylation of glycogen synthase kinase 3 beta, a kinase in the Wnt signaling pathway, was altered immediately following the binge cocaine paradigm and returned to normal levels 22 hours after the last cocaine injection. CONCLUSIONS Cocaine exposure during adolescence leads to molecular changes in the amygdala and decreases fear learning and anxiety in adulthood.
Collapse
Affiliation(s)
| | - Yolanda D. Black
- Department of Neurobiology and Behavior, University of California-Irvine, Irvine, California 92697
| | - Alipi V. Naydenov
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
| | - Fair R. Vassoler
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
| | - Ryan P. Hanlin
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
| | - Christine Konradi
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
- Center for Molecular Neuroscience, Vanderbilt University, Nashville, Tennessee, 37232
- Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee, 37203
| |
Collapse
|
19
|
Gutekunst CA, Stewart EN, Gross RE. Immunohistochemical Distribution of PlexinA4 in the Adult Rat Central Nervous System. Front Neuroanat 2010; 4. [PMID: 20700382 PMCID: PMC2914526 DOI: 10.3389/fnana.2010.00025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Accepted: 05/16/2010] [Indexed: 12/29/2022] Open
Abstract
PlexinA4 is the latest member to be identified of the PlexinA subfamily, critical transducers of class 3 semaphorin signaling as co-receptors to neuropilins 1 and 2. Despite functional information regarding the role of PlexinA4 in development and guidance of specific neuronal pathways, little is known about its distribution in the adult central nervous system (CNS). Here we report an in depth immunohistochemical analysis of PlexinA4 expression in the adult rat CNS. PlexinA4 staining was present in neurons and fibers throughout the brain and spinal cord, including neocortex, hippocampus, lateral hypothalamus, red nucleus, facial nucleus, and the mesencephalic trigeminal nucleus. PlexinA4 antibodies labeled fibers in the lateral septum, nucleus accumbens, several thalamic nuclei, substantia nigra pars reticulata, zona incerta, pontine reticular region, as well as in several cranial nerve nuclei. This constitutes the first detailed description of the topographic distribution of PlexinA4 in the adult CNS and will set the basis for future studies on the functional implications of PlexinA4 in adult brain physiology.
Collapse
|
20
|
Prasad AA, Pasterkamp RJ. Axon guidance in the dopamine system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 651:91-100. [PMID: 19731554 DOI: 10.1007/978-1-4419-0322-8_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Meso-diencephalic dopamine neurons (mdDA) neurons are located in the retrorubral field (RRF), substantia nigra pars compacta (SNc) and ventral tegmental area (VTA) and give rise to prominent ascending axon projections. These so-called mesotelencephalic projections are organized into three main pathways: the mesostriatal, mesocortical and mesolimbic pathways. Mesotelencephalic pathways in the adult nervous system have been studied in much detail as a result of their important physiological functions and their implication in psychiatric, neurological and neurodegenerative disease. In comparison, relatively little is known about the formation of these projection systems during embryonic and postnatal development. However, understanding the formation of mdDA neurons and their projections is essential for the design of effective therapies for mdDA neuron-associated neurological and neurodegenerative disorders. Here we summarize our current knowledge of the ontogeny of mdDA axon projections in subsystems of the developing rodent central nervous system (CNS) and discuss the cellular and molecular mechanisms that mediate mdDA axon guidance in these CNS regions.
Collapse
Affiliation(s)
- Asheeta A Prasad
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
21
|
Lull ME, Freeman WM, Vrana KE, Mash DC. Correlating human and animal studies of cocaine abuse and gene expression. Ann N Y Acad Sci 2008; 1141:58-75. [PMID: 18991951 DOI: 10.1196/annals.1441.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Gene expression changes resulting from cocaine abuse in both humans and animal models have been studied for several decades. Although human studies have been very useful at illuminating cocaine-related expression changes, there are many factors complicating these studies, including the difficulty of obtaining high-quality postmortem brain tissue and patient comorbidities. Animal models of cocaine abuse have served as valuable additions to human data and allow examination of specific aspects of cocaine abuse, including immediate early gene expression and the molecular effects of abstinence and relapse. In total, human and animal studies of cocaine abuse have uncovered gene expression changes in the brain related to a number of molecular functions, including the extracellular matrix, synaptic communication and neuroplasticity, receptors, ion channels and transporters, oligodendrocytes and myelin, apoptosis and cell death, mitochondrial function, signal transduction, and transcription factors. In addition, the mitogen-activated protein kinase and synaptic long-term potentiation signal transduction pathways are highlighted as pathways in which multiple components are altered by cocaine. Pathways and processes affected by changes in gene expression that overlap among multiple species may be promising pharmacotherapeutic targets for reducing the behavioral effects of cocaine abuse and the relapse potential observed in humans.
Collapse
Affiliation(s)
- Melinda E Lull
- Department of Pharmacology, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
22
|
EphrinB-EphB receptor signaling contributes to neuropathic pain by regulating neural excitability and spinal synaptic plasticity in rats. Pain 2008; 139:168-180. [DOI: 10.1016/j.pain.2008.03.019] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 03/17/2008] [Accepted: 03/18/2008] [Indexed: 11/20/2022]
|
23
|
Liu WT, Li HC, Song XS, Huang ZJ, Song XJ. EphB receptor signaling in mouse spinal cord contributes to physical dependence on morphine. FASEB J 2008; 23:90-8. [PMID: 18772347 DOI: 10.1096/fj.08-114462] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cellular and molecular mechanisms underlying opioid tolerance and dependence remain elusive. We investigated roles of EphB receptor tyrosine kinases--which play important roles in synaptic connection and plasticity during development and in the matured nervous system--in development and maintenance of physical dependence on morphine in the mouse spinal cord (SC). Spinal administration of an EphB receptor blocking reagent EphB2-Fc prevents and/or suppresses behavioral responses to morphine withdrawal and associated induction of c-Fos and depletion of calcitonin gene-related peptide. Western blotting and immunohistochemical fluorescence staining demonstrates that EphB1 receptor protein is significantly up-regulated in the spinal dorsal horn following escalating morphine treatment. Chronic morphine exposure and withdrawal significantly increased phosphorylation of N-methyl-D-aspartate receptor subunit NR2B as well as the activated forms of extracellular signal-regulated kinase and the cAMP response element binding protein in SC. The increased levels of phosphorylation of these molecules, however, are significantly inhibited by the EphB receptor blocker. These findings indicate that EphB receptor signaling, probably by interacting with NR2B in SC, contributes to the development of opioid physical dependence and withdrawal effects. This novel role for EphB receptor signaling suggests that these molecules may be useful therapeutic targets for preventing, minimizing, or reversing the development of opiate dependence.
Collapse
Affiliation(s)
- Wen-Tao Liu
- Department of Neurobiology, Parker University Research Institute, 2500 Walnut Hill Lane, Dallas, TX 75229, USA
| | | | | | | | | |
Collapse
|
24
|
Iwakura Y, Nawa H, Sora I, Chao MV. Dopamine D1 receptor-induced signaling through TrkB receptors in striatal neurons. J Biol Chem 2008; 283:15799-806. [PMID: 18381284 DOI: 10.1074/jbc.m801553200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In addition to its role as a neurotransmitter, dopamine can stimulate neurite outgrowth and morphological effects upon primary neurons. To investigate the signal transduction mechanisms used by dopamine in developing striatal neurons, we focused upon the effects of activating the dopamine D1 receptor. Using the D1 receptor agonist SKF38393, we found that Trk neurotrophin receptors were activated in embryonic day 18 striatal neurons. K-252a, a Trk tyrosine kinase inhibitor, and a dopamine D1 receptor antagonist could block the effects of SKF38393. The increase in TrkB phosphorylation was not the result of increased neurotrophin production. Induction of TrkB activity by SKF38393 was accompanied by the phosphorylation of several Trk signaling proteins, including phospholipase Cgamma, Akt, and MAPK. Biotinylation experiments followed by immunostaining by phospho-TrkB-specific antibodies indicated that the mechanism involved increased TrkB surface expression by dopamine D1 receptor activation. This increase in cell surface TrkB expression was dependent upon an increase in intracellular Ca(2+). These results indicate that stimulation of dopamine D1 receptors can be coupled to the neurotrophin receptor signaling to mediate the effects of dopamine upon striatal neurons.
Collapse
Affiliation(s)
- Yuriko Iwakura
- Molecular Neurobiology Program, Kimmel Center at Skirball Institute of Biomolecular Medicine, Department of Cell Biology, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | |
Collapse
|
25
|
Antipsychotic drugs alter neuronal development including ALM neuroblast migration and PLM axonal outgrowth in Caenorhabditis elegans. Int J Dev Neurosci 2008; 26:371-80. [PMID: 18282677 DOI: 10.1016/j.ijdevneu.2007.08.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 01/07/2008] [Accepted: 08/21/2007] [Indexed: 02/05/2023] Open
Abstract
Antipsychotic drugs are increasingly being prescribed for children and adolescents, and are used in pregnant women without a clear demonstration of safety in these populations. Global effects of these drugs on neurodevelopment (e.g., decreased brain size) have been reported in rats, but detailed knowledge about neuronal effects and mechanisms of action are lacking. Here we report on the evaluation of a comprehensive panel of antipsychotic drugs in a model organism (Caenorhabditis elegans) that is widely used to study neuronal development. Specifically, we examined the effects of the drugs on neuronal migration and axonal outgrowth in mechanosensory neurons visualized with green fluorescent protein expressed from the mec-3 promoter. Clozapine, fluphenazine, and haloperidol produced deficits in the development and migration of ALM neurons and axonal outgrowth in PLM neurons. The defects included failure of neuroblasts to migrate to the proper location, and excessive growth of axons past their normal termination point, together with abnormal morphological features of the processes. Although the antipsychotic drugs are potent antagonists of dopamine and serotonin receptors, the neurodevelopmental deficits were not rescued by co-incubation with serotonin or the dopaminergic agonist, quinpirole. Other antipsychotic drugs, risperidone, aripiprazole, quetiapine, trifluoperazine and olanzapine, also produced modest, but detectable, effects on neuronal development. This is the first report that antipsychotic drugs interfere with neuronal migration and axonal outgrowth in a developing nervous system.
Collapse
|
26
|
Numachi Y, Yoshida S, Yamashita M, Fujiyama K, Toda S, Matsuoka H, Kajii Y, Nishikawa T. Altered EphA5 mRNA expression in rat brain with a single methamphetamine treatment. Neurosci Lett 2007; 424:116-21. [PMID: 17714871 DOI: 10.1016/j.neulet.2007.07.025] [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] [Received: 05/25/2007] [Revised: 07/08/2007] [Accepted: 07/23/2007] [Indexed: 11/25/2022]
Abstract
Methamphetamine is a potent and indirect dopaminergic agonist which can cause chronic brain dysfunctions including drug abuse, drug dependence and drug-induced psychosis. Methamphetamine is known to trigger molecular mechanisms involved in associative learning and memory, and thereby alter patterns of synaptic connectivity. The persistent risk of relapse in methamphetamine abuse, dependence and psychosis may be caused by such alterations in synaptic connectivity. EphA5 receptors constitute large families of tyrosine kinase receptor and are expressed almost exclusively in the nervous system, especially in the limbic structures. Recent studies suggest EphA5 to be important in the topographic projection, development, and plasticity of limbic structures, and to be involved in dopaminergic neurotransmission. We used in situ hybridization to examine whether methamphetamine alters EphA5 mRNA expression in the brains of adult male Wister rats. EphA5 mRNA was widely distributed in the medial frontal cortex, cingulate cortex, piriform cortex, hippocampus, habenular nucleus and amygdala. Compared to baseline expression at 0h, EphA5 mRNA was significantly decreased (by 20%) in the medial frontal cortex at 24h, significantly increased (by 30%) in the amygdala at 9 and 24h, significantly but transiently decreased (by 30%) in the habenular nucleus at 1h after a single injection of methamphetamine. Methamphetamine did not change EphA5 mRNA expression in the cingulate cortex, piriform cortex or hippocampus. Our results that methamphetamine altered EphA5 mRNA expression in rat brain suggest methamphetamine could affect patterns of synaptic connectivity, which might be responsible for methamphetamine-induced chronic brain dysfunctions.
Collapse
Affiliation(s)
- Yohtaro Numachi
- Musashi Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chan ASL, Ng LWC, Poon LSW, Chan WWY, Wong YH. Dopaminergic and adrenergic toxicities on SK-N-MC human neuroblastoma cells are mediated through G protein signaling and oxidative stress. Apoptosis 2007; 12:167-79. [PMID: 17136323 DOI: 10.1007/s10495-006-0524-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dopamine and norepinephrine are neurotransmitters which participate in various regulatory functions of the human brain. These functions are lost in neurodegenerative diseases including Parkinson's disease and Alzheimer's disease. In this study, we used SK-N-MC neuroblastoma cells to investigate the cytotoxicities of high concentrations of dopamine and norepinephrine on neuronal cells. Dopamine, norepinephrine, as well as their corresponding synthetic agonists (SKF38393 and isoproterenol, respectively) triggered SK-N-MC cell death when applied at 50-100 muM persistently for 2 days. This catecholamine-induced cell death appears to be neuronal specific, as demonstrated by their inabilities of triggering apoptosis of A549 lung carcinoma cells and Cos-7 kidney fibroblasts. By pretreating SK-N-MC cells with target-specific inhibitors before administration of catecholamine, components of G protein signaling (i.e. G( s )/cAMP/PKA), monoamine oxidases, nitric oxide synthase, c-Jun N-terminal kinase and oxidative stress were found to be involved in this dopamine/norepinephrine-induced cytotoxicity, which subsequently led to caspase-dependent and -independent apoptotic responses as well as DNA degradation. In contrast, agonists of G( i )-coupled dopamine receptors and adrenergic receptors (quinpirole and UK14,304, respectively) were incapable of triggering apoptosis of SK-N-MC cells. Our results suggest that both G protein (G( s ))-mediated signaling cascade and oxidative stress participate in the dopamine/norepinephrine-induced neuronal apoptosis.
Collapse
Affiliation(s)
- Anthony S L Chan
- Department of Biochemistry, the Molecular Neuroscience Center, and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | | | | | | | | |
Collapse
|
28
|
Xie Z, Westmoreland SV, Bahn ME, Chen GL, Yang H, Vallender EJ, Yao WD, Madras BK, Miller GM. Rhesus monkey trace amine-associated receptor 1 signaling: enhancement by monoamine transporters and attenuation by the D2 autoreceptor in vitro. J Pharmacol Exp Ther 2007; 321:116-27. [PMID: 17234900 DOI: 10.1124/jpet.106.116863] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that directly responds to endogenous monoamines as well as amphetamine-related psychostimulants, including methamphetamine. In the present study, we demonstrate TAAR1 mRNA and protein expression in rhesus monkey brain regions associated with monoaminergic systems, variable cellular distribution of TAAR1 in rhesus monkey brain, and TAAR1 coexpression with the dopamine transporter (DAT) in a subset of dopamine neurons in both rhesus monkey and mouse substantia nigra. On this basis, we evaluated rhesus monkey TAAR1 activation by different compounds and its functional relation with monoamine transporters and the dopamine D2 receptor (D2) short isoform (D2s) autoreceptor in vitro using a cAMP response element-luciferase assay. TAAR1 activation by monoamines and amphetamine-related compounds was greatly enhanced by coexpression of dopamine, norepinephrine, or serotonin transporters, and the activation enhancement was blocked by monoamine transporter inhibitors. This enhancement did not occur in control experiments in which the dopamine D1 receptor (D1) was substituted for TAAR1. Furthermore, activation of TAAR1 by dopamine was completely inhibited by D2s when coexpressed with TAAR1, and this inhibition was blocked by the D2 antagonist raclopride. Last, dopamine activation of TAAR1 could induce c-FOS-luciferase expression but only in the presence of DAT, whereas dopamine activation of D1 resulted in equivalent c-FOS expression in the presence or absence of DAT. Together, these data reveal a broad agonist spectrum for TAAR1, a functional relation of TAAR1 with monoamine transporters and D2s, and a mechanism by which D2 receptor drugs can influence brain monoaminergic function and have efficacy through affecting TAAR1 signaling.
Collapse
MESH Headings
- Amphetamine/pharmacology
- Animals
- Autoreceptors/physiology
- Blotting, Western
- Cells, Cultured
- Central Nervous System Stimulants/pharmacology
- Dopamine Plasma Membrane Transport Proteins/physiology
- Electrophoresis, Polyacrylamide Gel
- Fluorescent Antibody Technique
- Genes, Reporter/physiology
- Immunohistochemistry
- Luciferases/metabolism
- Macaca mulatta
- Neurons/physiology
- Proto-Oncogene Proteins c-fos/physiology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Radioligand Assay
- Receptors, Dopamine D1/physiology
- Receptors, Dopamine D2/physiology
- Receptors, G-Protein-Coupled/biosynthesis
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/physiology
- Substantia Nigra/cytology
- Substantia Nigra/physiology
- Transfection
Collapse
Affiliation(s)
- Zhihua Xie
- Division of Neurochemistry, New England Primate Research Center, Harvard Medical School, One Pine Hill Dr., Southborough, MA 01772, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhang J, Vinuela A, Neely MH, Hallett PJ, Grant SGN, Miller GM, Isacson O, Caron MG, Yao WD. Inhibition of the dopamine D1 receptor signaling by PSD-95. J Biol Chem 2007; 282:15778-89. [PMID: 17369255 PMCID: PMC2649122 DOI: 10.1074/jbc.m611485200] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dopamine D1 receptors play an important role in movement, reward, and learning and are implicated in a number of neurological and psychiatric disorders. These receptors are concentrated in dendritic spines of neurons, including the spine head and the postsynaptic density. D1 within spines is thought to modulate the local channels and receptors to control the excitability and synaptic properties of spines. The molecular mechanisms mediating D1 trafficking, anchorage, and function in spines remain elusive. Here we show that the synaptic scaffolding protein PSD-95 thought to play a role in stabilizing glutamate receptors in the postsynaptic density, interacts with D1 and regulates its trafficking and function. Interestingly, the D1-PSD-95 interaction does not require the well characterized domains of PSD-95 but is mediated by the carboxyl-terminal tail of D1 and the NH(2) terminus of PSD-95, a region that is recognized only recently to participate in protein-protein interaction. Co-expression of PSD-95 with D1 in mammalian cells inhibits the D1-mediated cAMP accumulation without altering the total expression level or the agonist binding properties of the receptor. The diminished D1 signaling is mediated by reduced D1 expression at the cell surface as a consequence of an enhanced constitutive, dynamin-dependent endocytosis. In addition, genetically engineered mice lacking PSD-95 show a heightened behavioral response to either a D1 agonist or the psychostimulant amphetamine. These studies demonstrate a role for a glutamatergic scaffold in dopamine receptor signaling and trafficking and identify a new potential target for the modulation of abnormal dopaminergic function.
Collapse
Affiliation(s)
- Jingping Zhang
- Department of Psychiatry, Harvard Medical School, New England Primate Research Center, Southborough, Massachusetts 01772, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Xiao J, Gong S, LeDoux MS. Caytaxin deficiency disrupts signaling pathways in cerebellar cortex. Neuroscience 2006; 144:439-61. [PMID: 17092653 PMCID: PMC1868412 DOI: 10.1016/j.neuroscience.2006.09.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Revised: 09/15/2006] [Accepted: 09/20/2006] [Indexed: 11/28/2022]
Abstract
The genetically dystonic (dt) rat, an autosomal recessive model of generalized dystonia, harbors an insertional mutation in Atcay. As a result, dt rats are deficient in Atcay transcript and the neuronally-restricted protein caytaxin. Previous electrophysiological and biochemical studies have defined olivocerebellar pathways, particularly the climbing fiber projection to Purkinje cells, as sites of significant functional abnormality in dt rats. In normal rats, Atcay transcript is abundantly expressed in the granular and Purkinje cell layers of cerebellar cortex. To better understand the consequences of caytaxin deficiency in cerebellar cortex, differential gene expression was examined in dt rats and their normal littermates. Data from oligonucleotide microarrays and quantitative real-time reverse transcriptase-PCR (QRT-PCR) identified phosphatidylinositol signaling pathways, calcium homeostasis, and extracellular matrix interactions as domains of cellular dysfunction in dt rats. In dt rats, genes encoding the corticotropin-releasing hormone receptor 1 (CRH-R1, Crhr1) and plasma membrane calcium-dependent ATPase 4 (PMCA4, Atp2b4) showed the greatest up-regulation with QRT-PCR. Immunocytochemical experiments demonstrated that CRH-R1, CRH, and PMCA4 were up-regulated in cerebellar cortex of mutant rats. Along with previous electrophysiological and pharmacological studies, our data indicate that caytaxin plays a critical role in the molecular response of Purkinje cells to climbing fiber input. Caytaxin may also contribute to maturational events in cerebellar cortex.
Collapse
Affiliation(s)
| | | | - Mark S. LeDoux
- Address correspondence to: Mark S. LeDoux, M.D., Ph.D., University of Tennessee Health Science Center, Department of Neurology, 855 Monroe Avenue, Link Building-Suite 415, Memphis, Tennessee 38163, Phone: 901-448-1662, FAX: 901-448-7440,
| |
Collapse
|