1
|
Lee NC, Hsu PC, Liu YH, Wang HC, Chen TI, Chien YH, Hwu WL. Nigrostriatal tract defects in mice with aromatic l-amino acid decarboxylase deficiency. Neurobiol Dis 2024; 202:106707. [PMID: 39433135 DOI: 10.1016/j.nbd.2024.106707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
The development of the nigrostriatal dopaminergic (DA) pathway in the brain involves many transcriptional and chemotactic molecules, and a deficiency of these molecules can cause nigrostriatal tract defects. However, the role of the end product, dopamine, in nigrostriatal pathway development has not been described. In the present study, we analyzed a mouse model of congenital dopamine and serotonin deficiency, namely, the aromatic l-amino acid decarboxylase (AADC) deficiency (DdcKI) mouse model. We found via tyrosine hydroxylase (TH) immunofluorescence staining that the number of DA fibers in the stratum of 14-day-old DdcKI mice decreased. In TH-stained cleared whole brains of DdcKI mice, the numbers of DA neurons in the substantia nigra (SN) and the number of DA nerve bundles leaving the SN were both normal. However, we found that the nigrostriatal bundles in DdcKI mice were dispersed, taking aberrant routes to the striatum and spreading over a wide area. The total volume occupied by the nigrostriatal tract was increased, and the fraction of TH staining in the tract was decreased in DdcKI mice. Single-nucleus RNA sequencing analysis for mice 0, 7, and 14 days of age, revealed delayed axonogenesis and synapse formation in the striatum of DdcKI mice. The CellChat program inferred less cell-cell communication between striatal D1/D2 neurons but increased cell-cell communication involving neural precursors in DdcKI mice. Therefore, a congenital deficiency in dopamine affects nigrostriatal axon extension and striatal innervation. These nigrostriatal tract defects may limit the treatment efficacy for patients with TH or AADC deficiency.
Collapse
Affiliation(s)
- Ni-Chung Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Chun Hsu
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Han Liu
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hao-Chun Wang
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsu-I Chen
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Center for Precision Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
2
|
Haroon H, Ho AMC, Gupta VK, Dasari S, Sellgren CM, Cervenka S, Engberg G, Eren F, Erhardt S, Sung J, Choi DS. Cerebrospinal fluid proteomic signatures are associated with symptom severity of first-episode psychosis. J Psychiatr Res 2024; 171:306-315. [PMID: 38340697 PMCID: PMC10995989 DOI: 10.1016/j.jpsychires.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/04/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
Apart from their diagnostic, monitoring, or prognostic utility in clinical settings, molecular biomarkers may be instrumental in understanding the pathophysiology of psychiatric disorders, including schizophrenia. Using untargeted metabolomics, we recently identified eight cerebrospinal fluid (CSF) metabolites unique to first-episode psychosis (FEP) subjects compared to healthy controls (HC). In this study, we sought to investigate the CSF proteomic signatures associated with FEP. We employed 16-plex tandem mass tag (TMT) mass spectrometry (MS) to examine the relative protein abundance in CSF samples of 15 individuals diagnosed with FEP and 15 age-and-sex-matched healthy controls (HC). Multiple linear regression model (MLRM) identified 16 differentially abundant CSF proteins between FEP and HC at p < 0.01. Among them, the two most significant CSF proteins were collagen alpha-2 (IV) chain (COL4A2: standard mean difference [SMD] = -1.12, p = 1.64 × 10-4) and neuron-derived neurotrophic factor (NDNF: SMD = -1.03, p = 4.52 × 10-4) both of which were down-regulated in FEP subjects compared to HC. We also identified several potential CSF proteins associated with the pathophysiology and the symptom profile and severity in FEP subjects, including COL4A2, NDNF, hornerin (HRNR), contactin-6 (CNTN6), voltage-dependent calcium channel subunit alpha-2/delta-3 (CACNA2D3), tropomyosin alpha-3 chain (TPM3 and TPM4). Moreover, several protein signatures were associated with cognitive performance. Although the results need replication, our exploratory study suggests that CSF protein signatures can be used to increase the understanding of the pathophysiology of psychosis.
Collapse
Affiliation(s)
- Humza Haroon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Ada Man-Choi Ho
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Vinod K Gupta
- Division of Surgery Research, Department of Surgery, Rochester, MN, USA; Microbiome Program, Center for Individualized Medicine, Rochester, MN, USA
| | - Surendra Dasari
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden; Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Feride Eren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sophie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jaeyun Sung
- Division of Surgery Research, Department of Surgery, Rochester, MN, USA; Microbiome Program, Center for Individualized Medicine, Rochester, MN, USA; Division of Rheumatology, Department of Internal Medicine, Rochester, MN, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| |
Collapse
|
3
|
Cline MM, Juarez B, Hunker A, Regiarto EG, Hariadi B, Soden ME, Zweifel LS. Netrin-1 regulates the balance of synaptic glutamate signaling in the adult ventral tegmental area. eLife 2023; 12:e83760. [PMID: 36927614 PMCID: PMC10023152 DOI: 10.7554/elife.83760] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
The axonal guidance cue netrin-1 serves a critical role in neural circuit development by promoting growth cone motility, axonal branching, and synaptogenesis. Within the adult mouse brain, expression of the gene encoding (Ntn1) is highly enriched in the ventral midbrain where it is expressed in both GABAergic and dopaminergic neurons, but its function in these cell types in the adult system remains largely unknown. To address this, we performed viral-mediated, cell-type specific CRISPR-Cas9 mutagenesis of Ntn1 in the ventral tegmental area (VTA) of adult mice. Ntn1 loss-of-function in either cell type resulted in a significant reduction in excitatory postsynaptic connectivity. In dopamine neurons, the reduced excitatory tone had a minimal phenotypic behavioral outcome; however, reduced glutamatergic tone on VTA GABA neurons induced behaviors associated with a hyperdopaminergic phenotype. Simultaneous loss of Ntn1 function in both cell types largely rescued the phenotype observed in the GABA-only mutagenesis. These findings demonstrate an important role for Ntn1 in maintaining excitatory connectivity in the adult midbrain and that a balance in this connectivity within two of the major cell types of the VTA is critical for the proper functioning of the mesolimbic system.
Collapse
Affiliation(s)
- Marcella M Cline
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Molecular and Cellular Biology Program, University of WashingtonSeattleUnited States
| | - Barbara Juarez
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Avery Hunker
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Ernesto G Regiarto
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Bryan Hariadi
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Marta E Soden
- Department of Pharmacology, University of WashingtonSeattleUnited States
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Department of Pharmacology, University of WashingtonSeattleUnited States
| |
Collapse
|
4
|
Darcq E, Nouel D, Hernandez G, Pokinko M, Ash P, Moquin L, Gratton A, Kieffer B, Flores C. Reduced dopamine release in Dcc haploinsufficiency male mice abolishes the rewarding effects of cocaine but not those of morphine and ethanol. Psychopharmacology (Berl) 2023; 240:637-646. [PMID: 36471064 PMCID: PMC10296775 DOI: 10.1007/s00213-022-06288-1] [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: 09/20/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
RATIONALE The Netrin-1/DCC guidance cue pathway is critically involved in the adolescent organization of the mesocorticolimbic dopamine circuitry. Adult mice heterozygous for Dcc show reduced dopamine release in the nucleus accumbens in response to amphetamine and, in turn, blunted sensitivity to the rewarding effects of this drug. OBJECTIVE Here, we tested whether the protective effects of Dcc haploinsufficiency are specific to stimulant drugs of abuse or instead extrapolate to opioids and ethanol. METHODS We used the place preference paradigm to measure the rewarding effects of cocaine (20 mg/kg), morphine (5 or 10 mg/Kg), or ethanol (20%) in adult (PND 75) male Dcc haploinsufficient mice or their wild-type litter mates. In a second experiment, we compared in these two genotypes, in vivo dopamine release in the nucleus accumbens after a single i.p. injection of morphine (10 mg/kg). RESULTS We found reduced morphine-induced dopamine release in the nucleus accumbens of Dcc haploinsufficient male mice, but, contrary to the effects of stimulant drugs, there is no effect of genotype on morphine-induced conditioned preference. CONCLUSION These findings show that reduced drug-induced mesolimbic dopamine in Dcc haploinsufficient male mice protects specifically against the rewarding effects of stimulant drugs, but not against the rewarding properties of morphine and ethanol. These results suggest that these drugs exert their rewarding effect via different brain circuits.
Collapse
Affiliation(s)
- Emmanuel Darcq
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
- INSERM U1114, Centre de Recherche en Biomédecine de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Dominique Nouel
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | | | - Matthew Pokinko
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Integrated Program in Neuroscience (IPN), McGill University, Montréal, Québec, Canada
| | - Polina Ash
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Integrated Program in Neuroscience (IPN), McGill University, Montréal, Québec, Canada
| | - Luc Moquin
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | - Alain Gratton
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
| | - Brigitte Kieffer
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
- INSERM U1114, Centre de Recherche en Biomédecine de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montréal, Québec, Canada.
- Department of Psychiatry, McGill University, Montréal, Québec, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada.
- Department of Psychiatry, Douglas Mental Health University Institute, Perry Pavilion, Room 2111, 6875 LaSalle Boulevard, Montréal (Verdun), Québec, H4H 1R3, Canada.
| |
Collapse
|
5
|
Peters KZ, Naneix F. The role of dopamine and endocannabinoid systems in prefrontal cortex development: Adolescence as a critical period. Front Neural Circuits 2022; 16:939235. [PMID: 36389180 PMCID: PMC9663658 DOI: 10.3389/fncir.2022.939235] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/14/2022] [Indexed: 01/07/2023] Open
Abstract
The prefrontal cortex plays a central role in the control of complex cognitive processes including action control and decision making. It also shows a specific pattern of delayed maturation related to unique behavioral changes during adolescence and allows the development of adult cognitive processes. The adolescent brain is extremely plastic and critically vulnerable to external insults. Related to this vulnerability, adolescence is also associated with the emergence of numerous neuropsychiatric disorders involving alterations of prefrontal functions. Within prefrontal microcircuits, the dopamine and the endocannabinoid systems have widespread effects on adolescent-specific ontogenetic processes. In this review, we highlight recent advances in our understanding of the maturation of the dopamine system and the endocannabinoid system in the prefrontal cortex during adolescence. We discuss how they interact with GABA and glutamate neurons to modulate prefrontal circuits and how they can be altered by different environmental events leading to long-term neurobiological and behavioral changes at adulthood. Finally, we aim to identify several future research directions to help highlight gaps in our current knowledge on the maturation of these microcircuits.
Collapse
Affiliation(s)
- Kate Zara Peters
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom
| | - Fabien Naneix
- The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom,*Correspondence: Fabien Naneix
| |
Collapse
|
6
|
Heterozygous Dcc Mutant Mice Have a Subtle Locomotor Phenotype. eNeuro 2022; 9:ENEURO.0216-18.2021. [PMID: 35115383 PMCID: PMC8906791 DOI: 10.1523/eneuro.0216-18.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022] Open
Abstract
Axon guidance receptors such as deleted in colorectal cancer (DCC) contribute to the normal formation of neural circuits, and their mutations can be associated with neural defects. In humans, heterozygous mutations in DCC have been linked to congenital mirror movements, which are involuntary movements on one side of the body that mirror voluntary movements of the opposite side. In mice, obvious hopping phenotypes have been reported for bi-allelic Dcc mutations, while heterozygous mutants have not been closely examined. We hypothesized that a detailed characterization of Dcc heterozygous mice may reveal impaired corticospinal and spinal functions. Anterograde tracing of the Dcc+/− motor cortex revealed a normally projecting corticospinal tract, intracortical microstimulation (ICMS) evoked normal contralateral motor responses, and behavioral tests showed normal skilled forelimb coordination. Gait analyses also showed a normal locomotor pattern and rhythm in adult Dcc+/− mice during treadmill locomotion, except for a decreased occurrence of out-of-phase walk and an increased duty cycle of the stance phase at slow walking speed. Neonatal isolated Dcc+/− spinal cords had normal left-right and flexor-extensor coupling, along with normal locomotor pattern and rhythm, except for an increase in the flexor-related motoneuronal output. Although Dcc+/− mice do not exhibit any obvious bilateral impairments like those in humans, they exhibit subtle motor deficits during neonatal and adult locomotion.
Collapse
|
7
|
Lo PS, Rymar VV, Kennedy TE, Sadikot AF. The Netrin-1 Receptor DCC Promotes the Survival of a Subpopulation of Midbrain Dopaminergic Neurons: Relevance for Ageing and Parkinson's Disease. J Neurochem 2022; 161:254-265. [PMID: 35118677 PMCID: PMC9305203 DOI: 10.1111/jnc.15579] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/04/2022]
Abstract
Mechanisms that determine the survival of midbrain dopaminergic (mDA) neurons in the adult central nervous system (CNS) are not fully understood. Netrins are a family of secreted proteins that are essential for normal neural development. In the mature CNS, mDA neurons express particularly high levels of netrin‐1 and its receptor Deleted in Colorectal Cancer (DCC). Recent findings indicate that overexpressing netrin‐1 protects mDA neurons in animal models of Parkinson’s disease (PD), with a proposed pro‐apoptotic dependence function for DCC that triggers cell death in the absence of a ligand. Here, we sought to determine if DCC expression influences mDA neuron survival in young adult and ageing mice. To circumvent the perinatal lethality of DCC null mice, we selectively deleted DCC from mDA neurons utilizing DATcre/loxP gene‐targeting and examined neuronal survival in adult and aged animals. Reduced numbers of mDA neurons were detected in the substantia nigra pars compacta (SNc) of young adult DATcre/DCCfl/fl mice, with further reduction in aged DATcre/DCCfl/fl animals. In contrast to young adults, aged mice also exhibited a gene dosage effect, with fewer SNc mDA neurons in DCC heterozygotes (DATcre/DCCfl/wt). Notably, loss of mDA neurons in the SN was not uniform. Neuronal loss in the SN was limited to ventral tier mDA neurons, while mDA neurons in the dorsal tier of the SN, which resist degeneration in PD, were spared from the effect of DCC deletion in both young and aged mice. In the ventral tegmental area (VTA), young adult mice with conditional deletion of DCC had normal mDA neuronal numbers, while significant loss occurred in aged DATcre/DCCfl/fl and DATcre/DCCfl/wt mice compared to age‐matched wild‐type mice. Our results indicate that expression of DCC is required for the survival of subpopulations of mDA neurons and may be relevant to the degenerative processes in PD.![]()
Collapse
Affiliation(s)
- Pik-Shan Lo
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Qc, Canada.,Cone Laboratory, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Vladimir V Rymar
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Qc, Canada.,Cone Laboratory, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Timothy E Kennedy
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Qc, Canada
| | - Abbas F Sadikot
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Qc, Canada.,Cone Laboratory, Montreal Neurological Institute, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Reynolds LM, Flores C. Mesocorticolimbic Dopamine Pathways Across Adolescence: Diversity in Development. Front Neural Circuits 2021; 15:735625. [PMID: 34566584 PMCID: PMC8456011 DOI: 10.3389/fncir.2021.735625] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/17/2021] [Indexed: 12/26/2022] Open
Abstract
Mesocorticolimbic dopamine circuity undergoes a protracted maturation during adolescent life. Stable adult levels of behavioral functioning in reward, motivational, and cognitive domains are established as these pathways are refined, however, their extended developmental window also leaves them vulnerable to perturbation by environmental factors. In this review, we highlight recent advances in understanding the mechanisms underlying dopamine pathway development in the adolescent brain, and how the environment influences these processes to establish or disrupt neurocircuit diversity. We further integrate these recent studies into the larger historical framework of anatomical and neurochemical changes occurring during adolescence in the mesocorticolimbic dopamine system. While dopamine neuron heterogeneity is increasingly appreciated at molecular, physiological, and anatomical levels, we suggest that a developmental facet may play a key role in establishing vulnerability or resilience to environmental stimuli and experience in distinct dopamine circuits, shifting the balance between healthy brain development and susceptibility to psychiatric disease.
Collapse
Affiliation(s)
- Lauren M Reynolds
- Plasticité du Cerveau CNRS UMR8249, École supérieure de physique et de chimie industrielles de la Ville de Paris (ESPCI Paris), Paris, France.,Neuroscience Paris Seine CNRS UMR 8246 INSERM U1130, Institut de Biologie Paris Seine, Sorbonne Université, Paris, France
| | - Cecilia Flores
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| |
Collapse
|
9
|
Gao R, Peng X, Perry C, Sun H, Ntokou A, Ryu C, Gomez JL, Reeves BC, Walia A, Kaminski N, Neumark N, Ishikawa G, Black KE, Hariri LP, Moore MW, Gulati M, Homer RJ, Greif DM, Eltzschig HK, Herzog EL. Macrophage-derived netrin-1 drives adrenergic nerve-associated lung fibrosis. J Clin Invest 2021; 131:136542. [PMID: 33393489 PMCID: PMC7773383 DOI: 10.1172/jci136542] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is a macrophage-driven process of uncontrolled extracellular matrix accumulation. Neuronal guidance proteins such as netrin-1 promote inflammatory scarring. We found that macrophage-derived netrin-1 stimulates fibrosis through its neuronal guidance functions. In mice, fibrosis due to inhaled bleomycin engendered netrin-1-expressing macrophages and fibroblasts, remodeled adrenergic nerves, and augmented noradrenaline. Cell-specific knockout mice showed that collagen accumulation, fibrotic histology, and nerve-associated endpoints required netrin-1 of macrophage but not fibroblast origin. Adrenergic denervation; haploinsufficiency of netrin-1's receptor, deleted in colorectal carcinoma; and therapeutic α1 adrenoreceptor antagonism improved collagen content and histology. An idiopathic pulmonary fibrosis (IPF) lung microarray data set showed increased netrin-1 expression. IPF lung tissues were enriched for netrin-1+ macrophages and noradrenaline. A longitudinal IPF cohort showed improved survival in patients prescribed α1 adrenoreceptor blockade. This work showed that macrophages stimulate lung fibrosis via netrin-1-driven adrenergic processes and introduced α1 blockers as a potentially new fibrotic therapy.
Collapse
Affiliation(s)
- Ruijuan Gao
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xueyan Peng
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carrighan Perry
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Huanxing Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aglaia Ntokou
- Section of Cardiology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jose L. Gomez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Benjamin C. Reeves
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anjali Walia
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nir Neumark
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine, and
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meagan W. Moore
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mridu Gulati
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Robert J. Homer
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, and
| | - Daniel M. Greif
- Section of Cardiology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, University of Texas at Houston Medical School, Houston, Texas, USA
| | - Erica L. Herzog
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, and
| |
Collapse
|
10
|
Stratilov VA, Tyulkova EI, Vetrovoy OV. Prenatal Stress as a Factor of the
Development of Addictive States. J EVOL BIOCHEM PHYS+ 2020. [DOI: 10.1134/s0022093020060010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
García-Guillén IM, Alonso A, Morales-Delgado N, Andrés B, Puelles L, López-Bendito G, Marín F, Aroca P. Netrin-1/DCC Signaling Differentially Regulates the Migration of Pax7, Nkx6.1, Irx2, Otp, and Otx2 Cell Populations in the Developing Interpeduncular Nucleus. Front Cell Dev Biol 2020; 8:588851. [PMID: 33195252 PMCID: PMC7606981 DOI: 10.3389/fcell.2020.588851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/30/2020] [Indexed: 12/27/2022] Open
Abstract
The interpeduncular nucleus (IPN) is a hindbrain structure formed by three main subdivisions, the prodromal (Pro) domain located at the isthmus (Ist), and the rostral and caudal interpeduncular domains (IPR, IPC) within rhombomere 1 (r1). Various cell populations can be detected in the IPN through the expression of the Nkx6.1, Otp, Otx2, Pax7, and/or Irx2 transcription factors. These cell populations follow independent dorsoventral tangential and radial migratory routes targeting the ventral paramedian region of Ist and r1. Here we set out to examine the influence of the Netrin-1/DCC pathway on these migrations, since it is known to regulate other processes of neuronal migration in the brain. To this end, we analyzed IPN development in late gestational wild-type and DCC-/- mice, using mainly in situ hybridization (ISH) to identify the cells expressing each of the aforementioned genes. We found that the migration of Nkx6.1 + and Irx2 + cells into the Pro domain was strongly disrupted by the loss of DCC, as occurred with the migration of Pax7 +, Irx2 +, and Otp + cells that would normally form the IPR. In addition, there was mild impairment of the migration of the Pax7 + and Otx2 + cells that form the IPC. These results demonstrate that the Netrin-1/DCC signaling pathway is involved in the migration of most of the IPN populations, mainly affecting those of the Pro and IPR domains of this nucleus. There are psychiatric disorders that involve the medial habenula (mHb)-IPN system, so that this experimental model could provide a basis to study their neurodevelopmental etiology.
Collapse
Affiliation(s)
- Isabel M García-Guillén
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Antonia Alonso
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Nicanor Morales-Delgado
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain.,Department of Histology and Anatomy, School of Medicine, Miguel Hernández University, Alicante, Spain
| | - Belén Andrés
- Instituto de Neurociencias de Alicante, CSIC, Universidad Miguel Hernández, Alicante, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| | | | - Faustino Marín
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Pilar Aroca
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
12
|
Reynolds LM, Yetnikoff L, Pokinko M, Wodzinski M, Epelbaum JG, Lambert LC, Cossette MP, Arvanitogiannis A, Flores C. Early Adolescence is a Critical Period for the Maturation of Inhibitory Behavior. Cereb Cortex 2020; 29:3676-3686. [PMID: 30295713 DOI: 10.1093/cercor/bhy247] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/28/2018] [Accepted: 09/05/2018] [Indexed: 12/29/2022] Open
Abstract
Psychiatric conditions marked by impairments in cognitive control often emerge during adolescence, when the prefrontal cortex (PFC) and its inputs undergo structural and functional maturation and are vulnerable to disruption by external events. It is not known, however, whether there exists a specific temporal window within the broad range of adolescence when the development of PFC circuitry and its related behaviors are sensitive to disruption. Here we show, in male mice, that repeated exposure to amphetamine during early adolescence leads to impaired behavioral inhibition, aberrant PFC dopamine connectivity, and reduced PFC dopamine function in adulthood. Remarkably, these deficits are not observed following exposure to the exact same amphetamine regimen at later times. These findings demonstrate that there is a critical period for the disruption of the adolescent maturation of cognitive control and PFC dopamine function and suggest that early adolescence is particularly relevant to the emergence of psychopathology in humans.
Collapse
Affiliation(s)
- Lauren M Reynolds
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada.,Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Leora Yetnikoff
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, USA.,CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, USA
| | - Matthew Pokinko
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada.,Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Michael Wodzinski
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Julia G Epelbaum
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Laura C Lambert
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Marie-Pierre Cossette
- Department of Psychology, Center for Studies in Behavioural Neurobiology, Concordia University, Montréal, QC, Canada
| | - Andreas Arvanitogiannis
- Department of Psychology, Center for Studies in Behavioural Neurobiology, Concordia University, Montréal, QC, Canada
| | - Cecilia Flores
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| |
Collapse
|
13
|
Cuesta S, Nouel D, Reynolds LM, Morgunova A, Torres-Berrío A, White A, Hernandez G, Cooper HM, Flores C. Dopamine Axon Targeting in the Nucleus Accumbens in Adolescence Requires Netrin-1. Front Cell Dev Biol 2020; 8:487. [PMID: 32714924 PMCID: PMC7344302 DOI: 10.3389/fcell.2020.00487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
The fine arrangement of neuronal connectivity during development involves the coordinated action of guidance cues and their receptors. In adolescence, the dopamine circuitry is still developing, with mesolimbic dopamine axons undergoing target-recognition events in the nucleus accumbens (NAcc), while mesocortical projections continue to grow toward the prefrontal cortex (PFC) until adulthood. This segregation of mesolimbic versus mesocortical dopamine pathways is mediated by the guidance cue receptor DCC, which signals dopamine axons intended to innervate the NAcc to recognize this region as their final target. Whether DCC-dependent mesolimbic dopamine axon targeting in adolescence requires the action of its ligand, Netrin-1, is unknown. Here we combined shRNA strategies, quantitative analysis of pre- and post-synaptic markers of neuronal connectivity, and pharmacological manipulations to address this question. Similar to DCC levels in the ventral tegmental area, Netrin-1 expression in the NAcc is dynamic across postnatal life, transitioning from high to low expression across adolescence. Silencing Netrin-1 in the NAcc in adolescence results in an increase in the expanse of the dopamine input to the PFC in adulthood, with a corresponding increase in the number of presynaptic dopamine sites. This manipulation also results in altered dendritic spine density and morphology of medium spiny neurons in the NAcc in adulthood and in reduced sensitivity to the behavioral activating effects of the stimulant drug of abuse, amphetamine. These cellular and behavioral effects mirror those induced by Dcc haploinsufficiency within dopamine neurons in adolescence. Dopamine targeting in adolescence requires the complementary interaction between DCC receptors in mesolimbic dopamine axons and Netrin-1 in the NAcc. Factors regulating either DCC or Netrin-1 in adolescence can disrupt mesocorticolimbic dopamine development, rendering vulnerability or protection to phenotypes associated with psychiatric disorders.
Collapse
Affiliation(s)
- Santiago Cuesta
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Dominique Nouel
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Lauren M Reynolds
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Alice Morgunova
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Angélica Torres-Berrío
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Amanda White
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Giovanni Hernandez
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Cecilia Flores
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
14
|
Li HJ, Qu N, Hui L, Cai X, Zhang CY, Zhong BL, Zhang SF, Chen J, Xia B, Wang L, Jia QF, Li W, Chang H, Xiao X, Li M, Li Y. Further confirmation of netrin 1 receptor (DCC) as a depression risk gene via integrations of multi-omics data. Transl Psychiatry 2020; 10:98. [PMID: 32184385 PMCID: PMC7078234 DOI: 10.1038/s41398-020-0777-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/21/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies (GWAS) of major depression and its relevant biological phenotypes have been extensively conducted in large samples, and transcriptome-wide analyses in the tissues of brain regions relevant to pathogenesis of depression, e.g., dorsolateral prefrontal cortex (DLPFC), have also been widely performed recently. Integrating these multi-omics data will enable unveiling of depression risk genes and even underlying pathological mechanisms. Here, we employ summary data-based Mendelian randomization (SMR) and integrative risk gene selector (iRIGS) approaches to integrate multi-omics data from GWAS, DLPFC expression quantitative trait loci (eQTL) analyses and enhancer-promoter physical link studies to prioritize high-confidence risk genes for depression, followed by independent replications across distinct populations. These integrative analyses identify multiple high-confidence depression risk genes, and numerous lines of evidence supporting pivotal roles of the netrin 1 receptor (DCC) gene in this illness across different populations. Our subsequent explorative analyses further suggest that DCC significantly predicts neuroticism, well-being spectrum, cognitive function and putamen structure in general populations. Gene expression correlation and pathway analyses in DLPFC further show that DCC potentially participates in the biological processes and pathways underlying synaptic plasticity, axon guidance, circadian entrainment, as well as learning and long-term potentiation. These results are in agreement with the recent findings of this gene in neurodevelopment and psychiatric disorders, and we thus further confirm that DCC is an important susceptibility gene for depression, and might be a potential target for new antidepressants.
Collapse
Affiliation(s)
- Hui-Juan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Na Qu
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Li Hui
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xin Cai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Bao-Liang Zhong
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Shu-Fang Zhang
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Jing Chen
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Bin Xia
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qiu-Fang Jia
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wei Li
- Department of Blood Transfusion, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hong Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Yi Li
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China.
| |
Collapse
|
15
|
Vosberg DE, Leyton M, Flores C. The Netrin-1/DCC guidance system: dopamine pathway maturation and psychiatric disorders emerging in adolescence. Mol Psychiatry 2020; 25:297-307. [PMID: 31659271 PMCID: PMC6974431 DOI: 10.1038/s41380-019-0561-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/01/2019] [Accepted: 08/19/2019] [Indexed: 02/02/2023]
Abstract
Axon guidance molecules direct growing axons toward their targets, assembling the intricate wiring of the nervous system. One of these molecules, Netrin-1, and its receptor, DCC (deleted in colorectal cancer), has profound effects, in laboratory animals, on the adolescent expansion of mesocorticolimbic pathways, particularly dopamine. Now, a rapidly growing literature suggests that (1) these same alterations could occur in humans, and (2) genetic variants in Netrin-1 and DCC are associated with depression, schizophrenia, and substance use. Together, these findings provide compelling evidence that Netrin-1 and DCC influence mesocorticolimbic-related psychopathological states that emerge during adolescence.
Collapse
Affiliation(s)
- Daniel E Vosberg
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience (IPN), McGill University, Montreal, QC, Canada
- Population Neuroscience and Developmental Neuroimaging, Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Marco Leyton
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
- Integrated Program in Neuroscience (IPN), McGill University, Montreal, QC, Canada.
- Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| | - Cecilia Flores
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
- Integrated Program in Neuroscience (IPN), McGill University, Montreal, QC, Canada.
- Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Douglas Mental Health University Institute, Montreal, QC, Canada.
| |
Collapse
|
16
|
Kang DS, Kim IS, Baik JH, Kim D, Cocco L, Suh PG. The function of PLCγ1 in developing mouse mDA system. Adv Biol Regul 2019; 75:100654. [PMID: 31558431 DOI: 10.1016/j.jbior.2019.100654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 09/14/2019] [Accepted: 09/16/2019] [Indexed: 01/07/2023]
Abstract
During neural development, growing neuronal cells consistently sense and communicate with their surroundings through the use of signaling molecules. In this process, spatiotemporally well-coordinated intracellular signaling is a prerequisite for proper neuronal network formation. Thus, intense interest has focused on investigating the signaling mechanisms in neuronal structure formation that link the activation of receptors to the control of cell shape and motility. Recent studies suggest that Phospholipase C gamma1 (PLCγ1), a signal transducer, plays key roles in nervous system development by mediating specific ligand-receptor systems. In this overview of the most recent advances in the field, we discuss the mechanisms by which extracellular stimuli trigger PLCγ1 signaling and, the role PLCγ1 in nervous system development.
Collapse
Affiliation(s)
- Du-Seock Kang
- College of Life Science & Bioengineering, Korea Advanced Institute of Science & Technology (KAIST), Daejeon, South Korea.
| | - Il Shin Kim
- UNIST Central Research Facility, Ulsan National Institute of Science and Technology, South Korea.
| | - Ja-Hyun Baik
- Molecular Neurobiology Laboratory, Department of Life Sciences, Korea University, Seoul, 02841, South Korea.
| | - Daesoo Kim
- College of Life Science & Bioengineering, Korea Advanced Institute of Science & Technology (KAIST), Daejeon, South Korea.
| | - Lucio Cocco
- Cellular Signaling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, 689-798, South Korea; Korea Brain Research Institute, Daegu, 41062, South Korea.
| |
Collapse
|
17
|
Wong EW, Glasgow SD, Trigiani LJ, Chitsaz D, Rymar V, Sadikot A, Ruthazer ES, Hamel E, Kennedy TE. Spatial memory formation requires netrin-1 expression by neurons in the adult mammalian brain. ACTA ACUST UNITED AC 2019; 26:77-83. [PMID: 30770464 PMCID: PMC6380201 DOI: 10.1101/lm.049072.118] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 01/18/2019] [Indexed: 01/08/2023]
Abstract
Netrin-1 was initially characterized as an axon guidance molecule that is essential for normal embryonic neural development; however, many types of neurons continue to express netrin-1 in the postnatal and adult mammalian brain. Netrin-1 and the netrin receptor DCC are both enriched at synapses. In the adult hippocampus, activity-dependent secretion of netrin-1 by neurons potentiates glutamatergic synapse function, and is critical for long-term potentiation, an experimental cellular model of learning and memory. Here, we assessed the impact of neuronal expression of netrin-1 in the adult brain on behavior using tests of learning and memory. We show that adult mice exhibit impaired spatial memory following conditional deletion of netrin-1 from glutamatergic neurons in the hippocampus and neocortex. Further, we provide evidence that mice with conditional deletion of netrin-1 do not display aberrant anxiety-like phenotypes and show a reduction in self-grooming behavior. These findings reveal a critical role for netrin-1 expressed by neurons in the regulation of spatial memory formation.
Collapse
Affiliation(s)
- Edwin W Wong
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Stephen D Glasgow
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada.,NSERC CREATE Neuroengineering Training Program, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Lianne J Trigiani
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Daryan Chitsaz
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Vladimir Rymar
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Abbas Sadikot
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Edward S Ruthazer
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Edith Hamel
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Timothy E Kennedy
- Montréal Neurological Institute, Department of Neurology and Neurosurgery, 3801 Rue University, McGill University, Montréal, Québec H3A 2B4, Canada.,NSERC CREATE Neuroengineering Training Program, McGill University, Montréal, Québec H3A 2B4, Canada
| |
Collapse
|
18
|
Mesocorticolimbic Connectivity and Volumetric Alterations in DCC Mutation Carriers. J Neurosci 2018; 38:4655-4665. [PMID: 29712788 DOI: 10.1523/jneurosci.3251-17.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/29/2018] [Accepted: 04/07/2018] [Indexed: 01/25/2023] Open
Abstract
The axon guidance cue receptor DCC (deleted in colorectal cancer) plays a critical role in the organization of mesocorticolimbic pathways in rodents. To investigate whether this occurs in humans, we measured (1) anatomical connectivity between the substantia nigra/ventral tegmental area (SN/VTA) and forebrain targets, (2) striatal and cortical volumes, and (3) putatively associated traits and behaviors. To assess translatability, morphometric data were also collected in Dcc-haploinsufficient mice. The human volunteers were 20 DCC+/- mutation carriers, 16 DCC+/+ relatives, and 20 DCC+/+ unrelated healthy volunteers (UHVs; 28 females). The mice were 11 Dcc+/- and 16 wild-type C57BL/6J animals assessed during adolescence and adulthood. Compared with both control groups, the human DCC+/- carriers exhibited the following: (1) reduced anatomical connectivity from the SN/VTA to the ventral striatum [DCC+/+: p = 0.0005, r(effect size) = 0.60; UHV: p = 0.0029, r = 0.48] and ventral medial prefrontal cortex (DCC+/+: p = 0.0031, r = 0.53; UHV: p = 0.034, r = 0.35); (2) lower novelty-seeking scores (DCC+/+: p = 0.034, d = 0.82; UHV: p = 0.019, d = 0.84); and (3) reduced striatal volume (DCC+/+: p = 0.0009, d = 1.37; UHV: p = 0.0054, d = 0.93). Striatal volumetric reductions were also present in Dcc+/- mice, and these were seen during adolescence (p = 0.0058, d = 1.09) and adulthood (p = 0.003, d = 1.26). Together these findings provide the first evidence in humans that an axon guidance gene is involved in the formation of mesocorticolimbic circuitry and related behavioral traits, providing mechanisms through which DCC mutations might affect susceptibility to diverse neuropsychiatric disorders.SIGNIFICANCE STATEMENT Opportunities to study the effects of axon guidance molecules on human brain development have been rare. Here, the identification of a large four-generational family that carries a mutation to the axon guidance molecule receptor gene, DCC, enabled us to demonstrate effects on mesocorticolimbic anatomical connectivity, striatal volumes, and personality traits. Reductions in striatal volumes were replicated in DCC-haploinsufficient mice. Together, these processes might influence mesocorticolimbic function and susceptibility to diverse neuropsychiatric disorders.
Collapse
|
19
|
Kelaï S, Ramoz N, Moalic JM, Noble F, Mechawar N, Imbeaud S, Turecki G, Simonneau M, Gorwood P, Maussion G. Netrin G1: its downregulation in the nucleus accumbens of cocaine-conditioned mice and genetic association in human cocaine dependence. Addict Biol 2018; 23:448-460. [PMID: 28074533 DOI: 10.1111/adb.12485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/03/2016] [Accepted: 11/22/2016] [Indexed: 12/17/2022]
Abstract
Netrin G1 is a presynaptic ligand involved in axonal projection. Although molecular mechanisms underlying cocaine addiction are still poorly understood, Netrin G1 might have a role as a regulator of anxiety, fear and spatial memory, behavioural traits impaired in the context of cocaine exposure. In this study, the Netrin G1 (Ntng1) expression was investigated in the nucleus accumbens of mice primarily conditioned to cocaine using a place preference paradigm. A genetic association study was then conducted on 146 multiplex families of the Collaborative study on Genetics of Alcoholism, in which seven single nucleotide polymorphisms located in the NTNG1 gene were genotyped. NTNG1 expression levels were also quantified in BA10, BA46 and the cerebellum of healthy controls (with no Axis 1 psychopathology). Decreased Ntng1 expression was initially observed in the nucleus accumbens of mice conditioned to cocaine. Significant genetic family-based associations were detected between NTNG1 polymorphisms and cocaine dependence. NTNG1 expression in BA10, BA46 and the cerebellum, however, were not significantly associated with any allele or haplotype of this gene. These results confirm that Ntng1 expression is disturbed in the nucleus accumbens of mice, after cocaine conditioning. A haplotype of NTNG1 was found to constitute a vulnerability factor for cocaine use disorder in patients, although none of its single nucleotide polymorphisms were associated with a differential expression pattern in healthy controls. The data suggest that change in the Ntng1 expression is a consequence of cocaine exposure, and that some of its genetic markers are associated with a greater risk for cocaine use disorder.
Collapse
Affiliation(s)
- Sabah Kelaï
- INSERM U894, Centre de Psychiatrie & Neurosciences; University Paris Descartes; Paris France
| | - Nicolas Ramoz
- INSERM U894, Centre de Psychiatrie & Neurosciences; University Paris Descartes; Paris France
| | - Jean-Marie Moalic
- INSERM U894, Centre de Psychiatrie & Neurosciences; University Paris Descartes; Paris France
| | - Florence Noble
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche; France
- Institut national de la santé et de la recherche médicale; Paris France
- Université Paris Descartes, Laboratoire de Neuropsychopharmacologie des Addictions; France
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute; McGill University; Canada
| | - Sandrine Imbeaud
- Centre de Génétique Moléculaire, FRE 3144, CNRS and Gif/Orsay DNA Microarray Platform (GODMAP); France
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute; McGill University; Canada
| | - Michel Simonneau
- INSERM U894, Centre de Psychiatrie & Neurosciences; University Paris Descartes; Paris France
| | - Philip Gorwood
- INSERM U894, Centre de Psychiatrie & Neurosciences; University Paris Descartes; Paris France
- Hôpital Sainte-Anne (CMME); University Paris Descartes; France
| | - Gilles Maussion
- INSERM U894, Centre de Psychiatrie & Neurosciences; University Paris Descartes; Paris France
- McGill Group for Suicide Studies, Douglas Mental Health University Institute; McGill University; Canada
| |
Collapse
|
20
|
Making Dopamine Connections in Adolescence. Trends Neurosci 2017; 40:709-719. [PMID: 29032842 DOI: 10.1016/j.tins.2017.09.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/07/2017] [Accepted: 09/14/2017] [Indexed: 12/13/2022]
Abstract
A dramatic maturational process ongoing in adolescence is prefrontal cortex development, including its dopamine innervation. Dopamine axons grow from the striatum to the prefrontal cortex, the only known case of long-distance axon growth during adolescence. This is coordinated by the Netrin-1 guidance cue receptor DCC (deleted in colorectal cancer), which in turn controls the intrinsic development of the prefrontal cortex itself. Stimulant drugs in adolescence alter DCC in dopamine neurons and, in turn prefrontal cortex maturation, impacting cognitive abilities. Variations in DCC expression are linked to psychiatric conditions of prefrontal cortex dysfunction, and microRNA regulation of DCC may be key to determining adolescent vulnerability or resilience. Since early interventions are proving to effectively ameliorate disease outcome, the Netrin-1 system is a promising therapeutic target.
Collapse
|
21
|
Francardo V, Schmitz Y, Sulzer D, Cenci MA. Neuroprotection and neurorestoration as experimental therapeutics for Parkinson's disease. Exp Neurol 2017; 298:137-147. [PMID: 28988910 DOI: 10.1016/j.expneurol.2017.10.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/25/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022]
Abstract
Disease-modifying treatments remain an unmet medical need in Parkinson's disease (PD). Such treatments can be operationally defined as interventions that slow down the clinical evolution to advanced disease milestones. A treatment may achieve this outcome by either inhibiting primary neurodegenerative events ("neuroprotection") or boosting compensatory and regenerative mechanisms in the brain ("neurorestoration"). Here we review experimental paradigms that are currently used to assess the neuroprotective and neurorestorative potential of candidate treatments in animal models of PD. We review some key molecular mediators of neuroprotection and neurorestoration in the nigrostriatal dopamine pathway that are likely to exert beneficial effects on multiple neural systems affected in PD. We further review past and current strategies to therapeutically stimulate these mediators, and discuss the preclinical evidence that exercise training can have neuroprotective and neurorestorative effects. A future translational task will be to combine behavioral and pharmacological interventions to exploit endogenous mechanisms of neuroprotection and neurorestoration for therapeutic purposes. This type of approach is likely to provide benefit to many PD patients, despite the clinical, etiological, and genetic heterogeneity of the disease.
Collapse
Affiliation(s)
- Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Yvonne Schmitz
- Departments Neurology, Psychiatry, Pharmacology, Columbia University Medical Center: Division of Molecular Therapeutics, New York State Psychiatric Institute, New York 10032, NY, USA
| | - David Sulzer
- Departments Neurology, Psychiatry, Pharmacology, Columbia University Medical Center: Division of Molecular Therapeutics, New York State Psychiatric Institute, New York 10032, NY, USA
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
22
|
Brignani S, Pasterkamp RJ. Neuronal Subset-Specific Migration and Axonal Wiring Mechanisms in the Developing Midbrain Dopamine System. Front Neuroanat 2017; 11:55. [PMID: 28740464 PMCID: PMC5502286 DOI: 10.3389/fnana.2017.00055] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/20/2017] [Indexed: 01/01/2023] Open
Abstract
The midbrain dopamine (mDA) system is involved in the control of cognitive and motor behaviors, and is associated with several psychiatric and neurodegenerative diseases. mDA neurons receive diverse afferent inputs and establish efferent connections with many brain areas. Recent studies have unveiled a high level of molecular and cellular heterogeneity within the mDA system with specific subsets of mDA neurons displaying select molecular profiles and connectivity patterns. During mDA neuron development, molecular differences between mDA neuron subsets allow the establishment of subset-specific afferent and efferent connections and functional roles. In this review, we summarize and discuss recent work defining novel mDA neuron subsets based on specific molecular signatures. Then, molecular cues are highlighted that control mDA neuron migration during embryonic development and that facilitate the formation of selective patterns of efferent connections. The review focuses largely on studies that show differences in these mechanisms between different subsets of mDA neurons and for which in vivo data is available, and is concluded by a section that discusses open questions and provides directions for further research.
Collapse
Affiliation(s)
- Sara Brignani
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands
| | - R J Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands
| |
Collapse
|
23
|
Gore BB, Miller SM, Jo YS, Baird MA, Hoon M, Sanford CA, Hunker A, Lu W, Wong RO, Zweifel LS. Roundabout receptor 2 maintains inhibitory control of the adult midbrain. eLife 2017; 6. [PMID: 28394253 PMCID: PMC5419739 DOI: 10.7554/elife.23858] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/09/2017] [Indexed: 12/22/2022] Open
Abstract
The maintenance of excitatory and inhibitory balance in the brain is essential for its function. Here we find that the developmental axon guidance receptor Roundabout 2 (Robo2) is critical for the maintenance of inhibitory synapses in the adult ventral tegmental area (VTA), a brain region important for the production of the neurotransmitter dopamine. Following selective genetic inactivation of Robo2 in the adult VTA of mice, reduced inhibitory control results in altered neural activity patterns, enhanced phasic dopamine release, behavioral hyperactivity, associative learning deficits, and a paradoxical inversion of psychostimulant responses. These behavioral phenotypes could be phenocopied by selective inactivation of synaptic transmission from local GABAergic neurons of the VTA, demonstrating an important function for Robo2 in regulating the excitatory and inhibitory balance of the adult brain. DOI:http://dx.doi.org/10.7554/eLife.23858.001 Although no two people are alike, we all share the same basic brain structure. This similarity arises because the same developmental program takes place in every human embryo. Specific genes are activated in a designated sequence to generate the structure of a typical human brain. But what happens to these genes when development is complete – do they remain active in the adult brain? A gene known as Robo2 encodes a protein that helps neurons find their way through the developing brain. Many of these neurons will ultimately form part of the brain’s reward system. This is a network of brain regions that communicate with one another using a chemical called dopamine. The reward system contributes to motivation, learning and memory, and also underlies drug addiction. In certain mental illnesses such as Parkinson’s disease and schizophrenia, the dopamine-producing neurons in the reward system work incorrectly or die. To find out whether Robo2 is active in the mature nervous system, Gore et al. used genetic techniques to selectively remove the gene from the reward system of adult mice. Doing so reduced the ability of the dopamine neurons within the reward system to inhibit one another, which in turn increased their activity. This changed the behavior of the mice, making them hyperactive and less able to learn and remember. Cocaine makes normal mice more active; however, mice that lacked the Robo2 gene became less active when given cocaine. Overall, the work of Gore et al. suggests that developmental axon guidance genes remain important in the adult brain. Studying developmental genes such as Robo2 may therefore open up new treatment possibilities for a number of mental illnesses and brain disorders. DOI:http://dx.doi.org/10.7554/eLife.23858.002
Collapse
Affiliation(s)
- Bryan B Gore
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, United States.,Department of Pharmacology, University of Washington, Seattle, United States
| | - Samara M Miller
- Department of Pharmacology, University of Washington, Seattle, United States
| | - Yong Sang Jo
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, United States.,Department of Pharmacology, University of Washington, Seattle, United States
| | - Madison A Baird
- Department of Pharmacology, University of Washington, Seattle, United States
| | - Mrinalini Hoon
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Christina A Sanford
- Department of Pharmacology, University of Washington, Seattle, United States
| | - Avery Hunker
- Department of Pharmacology, University of Washington, Seattle, United States
| | - Weining Lu
- Department of Medicine, Renal Section, Boston University Medical Center, Boston, United States
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, United States.,Department of Pharmacology, University of Washington, Seattle, United States
| |
Collapse
|
24
|
Li Y, Qiao X, Yin F, Guo H, Huang X, Lai J, Wei S. A Population-Based Study of Four Genes Associated with Heroin Addiction in Han Chinese. PLoS One 2016; 11:e0163668. [PMID: 27676367 PMCID: PMC5038970 DOI: 10.1371/journal.pone.0163668] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/11/2016] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that variants in FAT atypical cadherin 3 (FAT3), kinectin 1 (KTN1), discs large homolog2 (DLG2) and deleted in colorectal cancer (DCC) genes influence the structure of the human mesolimbic reward system. We conducted a systematic analysis of the potential functional single nucleotide polymorphisms (SNPs) in these genes associated with heroin addiction. We scanned the functional regions of these genes and identified 20 SNPs for genotyping by using the SNaPshot method. A total of 1080 samples, comprising 523 cases and 557 controls, were analyzed. We observed that DCC rs16956878, rs12607853, and rs2292043 were associated with heroin addiction. The T alleles of rs16956878 (p = 0.0004) and rs12607853 (p = 0.002) were significantly enriched in the case group compared with the controls. A lower incidence of the C allele of rs2292043 (p = 0.002) was observed in the case group. In block 2 of DCC (rs2292043-rs12607853-rs16956878), the frequency of the T-T-T haplotype was significantly higher in the case group than in the control group (p = 0.024), and fewer C-C-C haplotypes (p = 0.006) were detected in the case group. DCC may be an important candidate gene in heroin addiction, and rs16956878, rs12607853, and rs2292043 may be risk factors, thereby providing a basis for further genetic and biological research.
Collapse
Affiliation(s)
- Yunxiao Li
- College of Forensic Science, Xi’an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi’an, PR China
| | - Xiaomeng Qiao
- College of Forensic Science, Xi’an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi’an, PR China
| | - Fangyuan Yin
- College of Forensic Science, Xi’an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi’an, PR China
| | - Hao Guo
- College of Forensic Science, Xi’an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi’an, PR China
| | - Xin Huang
- College of Forensic Science, Xi’an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi’an, PR China
| | - Jianghua Lai
- College of Forensic Science, Xi’an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi’an, PR China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, PR China
| | - Shuguang Wei
- College of Forensic Science, Xi’an Jiaotong University, Key Laboratory of Ministry of Public Health for Forensic Science, Xi’an, PR China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, PR China
- * E-mail:
| |
Collapse
|
25
|
Mesocortical Dopamine Phenotypes in Mice Lacking the Sonic Hedgehog Receptor Cdon. eNeuro 2016; 3:eN-NWR-0009-16. [PMID: 27419218 PMCID: PMC4942720 DOI: 10.1523/eneuro.0009-16.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/21/2022] Open
Abstract
Motivated behaviors and many psychopathologies typically involve changes in dopamine release from the projections of the ventral tegmental area (VTA) and/or the substantia nigra pars compacta (SNc). The morphogen Sonic Hedgehog (Shh) specifies fates of midbrain dopamine neurons, but VTA-specific effects of Shh signaling are also being uncovered. In this study, we assessed the role of the Shh receptor Cdon in the development of VTA and SNc dopamine neurons. We find that Cdon is expressed in the proliferating progenitor zone of the embryonic ventral midbrain and that the number of proliferating cells in this region is increased in mouse Cdon(-/-) embryos. Consistent with a role of Shh in the regulation of neuronal proliferation in this region, we find that the number of tyrosine hydroxylase (TH)-positive neurons is increased in the VTA of Cdon(-/-) mice at birth and that this effect endures into adulthood. In contrast, the number of TH-positive neurons in the SNc is not altered in Cdon(-/-) mice at either age. Moreover, adult Cdon(-/-) mice have a greater number of medial prefrontal cortex (mPFC) dopamine presynaptic sites, and increased baseline concentrations of dopamine and dopamine metabolites selectively in this region. Finally, consistent with increased dopamine function in the mPFC, we find that adult Cdon(-/-) mice fail to exhibit behavioral plasticity upon repeated amphetamine treatment. Based on these data, we suggest that Cdon plays an important role encoding the diversity of dopamine neurons in the midbrain, influencing both the development of the mesocortical dopamine pathway and behavioral outputs that involve this neural circuitry.
Collapse
|
26
|
An Association Study Between Genetic Polymorphisms in Functional Regions of Five Genes and the Risk of Schizophrenia. J Mol Neurosci 2016; 59:366-75. [PMID: 27055860 DOI: 10.1007/s12031-016-0751-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/28/2016] [Indexed: 02/08/2023]
Abstract
Schizophrenia is a severe mental disorder that is likely to be strongly determined by genetic factors. To identify markers of disks, large homolog 2 (DLG2), FAT atypical cadherin 3 (FAT3), kinectin1 (KTN1), deleted in colorectal carcinoma (DCC), and glycogen synthase kinase-3β (GSK3β) that contribute to the genetic susceptibility to schizophrenia, we systematically screened for polymorphisms in the functional regions of these genes. A total of 22 functional single-nucleotide polymorphisms (SNPs) in 940 Chinese subjects were genotyped using SNaPshot. The results first suggested that the allelic and genotypic frequencies of the DCC polymorphism rs2229080 were nominally associated with schizophrenia. The patients were significantly less likely to be CC homozygous (P = 0.005, odds ratio [OR] = 0.635, 95 % confidence interval [95 % CI] = 0.462-0.873), and the schizophrenia subjects exhibited lower frequency of the C allele (P = 0.024, OR = 0.811, 95 % CI = 0.676-0.972). Regarding GSK3β, there was a significant difference in genotype distribution of rs3755557 between schizophrenia and healthy control subjects (P = 0.009). The patients exhibited a significantly lower frequency of the T allele of rs3755557 (P = 0.002, OR = 0.654, 95 % CI = 0.498-0.860). Our results point to the polymorphisms of DCC and GSK3β as contributors to the genetic basis of individual differences in the susceptibility to schizophrenia.
Collapse
|
27
|
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: 23] [Impact Index Per Article: 2.6] [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
|
28
|
Bissonette GB, Roesch MR. Development and function of the midbrain dopamine system: what we know and what we need to. GENES BRAIN AND BEHAVIOR 2015; 15:62-73. [PMID: 26548362 DOI: 10.1111/gbb.12257] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/11/2015] [Accepted: 10/01/2015] [Indexed: 01/29/2023]
Abstract
The past two decades have seen an explosion in our understanding of the origin and development of the midbrain dopamine system. Much of this work has been focused on the aspects of dopamine neuron development related to the onset of movement disorders such as Parkinson's disease, with the intent of hopefully delaying, preventing or fixing symptoms. While midbrain dopamine degeneration is a major focus for treatment and research, many other human disorders are impacted by abnormal dopamine, including drug addiction, autism and schizophrenia. Understanding dopamine neuron ontogeny and how dopamine connections and circuitry develops may provide us with key insights into potentially important avenues of research for other dopamine-related disorders. This review will provide a brief overview of the major molecular and genetic players throughout the development of midbrain dopamine neurons and what we know about the behavioral- and disease-related implications associated with perturbations to midbrain dopamine neuron development. We intend to combine the knowledge of two broad fields of neuroscience, both developmental and behavioral, with the intent on fostering greater discussion between branches of neuroscience in the service of addressing complex cognitive questions from a developmental perspective and identifying important gaps in our knowledge for future study.
Collapse
Affiliation(s)
- G B Bissonette
- Department of Psychology, University of Maryland, College Park, MD, USA.,Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA
| | - M R Roesch
- Department of Psychology, University of Maryland, College Park, MD, USA.,Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA
| |
Collapse
|
29
|
Pokinko M, Moquin L, Torres-Berrío A, Gratton A, Flores C. Resilience to amphetamine in mouse models of netrin-1 haploinsufficiency: role of mesocortical dopamine. Psychopharmacology (Berl) 2015; 232:3719-29. [PMID: 26264903 DOI: 10.1007/s00213-015-4032-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/13/2015] [Indexed: 11/25/2022]
Abstract
RATIONALE Signaling through the netrin-1 receptor, deleted in colorectal cancer (DCC), in dopamine neurons controls the extent of their innervation to the medial prefrontal cortex (mPFC) during adolescence. In mice, dcc haploinsufficiency results in increased mPFC dopamine innervation and concentrations in adulthood. In turn, dcc haploinsufficiency leads to resilience to the effects of stimulant drugs of abuse on dopamine release in the nucleus accumbens and behavior. OBJECTIVES First, we set out to determine whether increased mPFC dopamine innervation causes blunted behavioral responses to amphetamine in adult dcc haploinsufficient mice. Second, we investigated whether unc5c, another netrin-1 receptor expressed by dopamine neurons, is involved in these effects. Third, we assessed whether haploinsufficiency of netrin-1 itself leads to blunted behavioral responding to amphetamine, whether this phenotype emerges before or after adolescence and whether increased mPFC dopamine input is the underlying mechanism. RESULTS Adult, but not adolescent, dcc, unc5c and netrin-1 haploinsufficient mice exhibit blunted behavioral responses to amphetamine. Furthermore, adult dcc, unc5c, and netrin-1 haploinsufficient mice have exaggerated mPFC dopamine concentrations in comparison to their wild-type littermates. Importantly, resilience to amphetamine-induced behavioral activation in all the three mouse models is abolished by selective dopamine depletion in the medial prefrontal cortex. CONCLUSIONS dcc, unc5c, or netrin-1 haploinsufficiency leads to increased dopamine content in the mPFC and to resilience against amphetamine-induced behavioral activation. Our findings raise the hypothesis that DCC, UNC5C, and netrin-1 act in concert to organize the adolescent development of mesocortical dopamine innervation and, in turn, determine behavioral responses to drugs of abuse.
Collapse
Affiliation(s)
- Matthew Pokinko
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | | | | | | | | |
Collapse
|
30
|
Abstract
ABSTRACT
Midbrain dopaminergic (mDA) neuron development has been an intense area of research during recent years. This is due in part to a growing interest in regenerative medicine and the hope that treatment for diseases affecting mDA neurons, such as Parkinson's disease (PD), might be facilitated by a better understanding of how these neurons are specified, differentiated and maintained in vivo. This knowledge might help to instruct efforts to generate mDA neurons in vitro, which holds promise not only for cell replacement therapy, but also for disease modeling and drug discovery. In this Primer, we will focus on recent developments in understanding the molecular mechanisms that regulate the development of mDA neurons in vivo, and how they have been used to generate human mDA neurons in vitro from pluripotent stem cells or from somatic cells via direct reprogramming. Current challenges and future avenues in the development of a regenerative medicine for PD will be identified and discussed.
Collapse
Affiliation(s)
- Ernest Arenas
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Mark Denham
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus 8000, Denmark
| | - J. Carlos Villaescusa
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno 61137, Czech Republic
| |
Collapse
|
31
|
Abstract
The mesofrontal dopaminergic circuit, which connects the midbrain motivation center to the cortical executive center, is engaged in control of motivated behaviors. In addition, deficiencies in this circuit are associated with adolescent-onset psychiatric disorders in humans. Developmental studies suggest that the mesofrontal circuit exhibits a protracted maturation through adolescence. However, whether the structure and function of this circuit are modifiable by activity in dopaminergic neurons during adolescence remains unknown. Using optogenetic stimulation and in vivo two-photon imaging in adolescent mice, we found that phasic, but not tonic, dopamine neuron activity induces the formation of mesofrontal axonal boutons. In contrast, in adult mice, the effect of phasic activity diminishes. Furthermore, our results showed that dopaminergic and glutamatergic transmission regulate this axonal plasticity in adolescence and inhibition of dopamine D2-type receptors restores this plasticity in adulthood. Finally, we found that phasic activation of dopamine neurons also induces greater changes in mesofrontal circuit activity and psychomotor response in adolescent mice than in adult mice. Together, our findings demonstrate that the structure and function of the mesofrontal circuit are modifiable by phasic activity in dopaminergic neurons during adolescence and suggest that the greater plasticity in adolescence may facilitate activity-dependent strengthening of dopaminergic input and improvement in behavioral control.
Collapse
|
32
|
Haloperidol treatment downregulates DCC expression in the ventral tegmental area. Neurosci Lett 2014; 575:58-62. [PMID: 24861518 DOI: 10.1016/j.neulet.2014.05.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/26/2014] [Accepted: 05/14/2014] [Indexed: 11/23/2022]
Abstract
A core feature in the pathophysiology of schizophrenia is abnormal development and function of mesocorticolimbic dopamine (DA) circuitry. We have previously shown that variations in the function of the netrin-1 receptor, deleted in colorectal cancer (DCC), result in changes to the development, organization and ongoing plasticity of DA circuitry. In rodents, repeated exposure to the indirect DA-agonist, amphetamine upregulates DCC expression in the ventral tegmental area (VTA), but not in DA terminal regions. This elevation in DCC expression is associated with increased vulnerability to developing and maintaining sensitized mesolimbic DA function. Antipsychotic medications remain the best treatment option for managing the symptoms in schizophrenia. The peak effects of these medications are gradual, suggesting that a therapeutic component of antipsychotic treatment involves structural reorganization. Here we assessed whether repeated exposure to typical and atypical antipsychotics could also regulate DCC. Adult mice were orally administered haloperidol, clozapine, or risperidone via their drinking water for 4 weeks. Levels of DCC were measured by Western blot analysis of tissue punches of the VTA, medial prefrontal cortex, nucleus accumbens, and dorsal striatum. Haloperidol decreased DCC levels by approximately 50% in the VTA, but not in DA targets. Furthermore, haloperidol did not alter UNC-5 homologue levels, another family of netrin-1 receptors, confirming that its effects target DCC-mediated netrin-1 signaling specifically. The atypical antipsychotics did not alter DCC expression. These results suggest that typical antipsychotics induce selective functional reorganization in the VTA via DCC-mediated netrin-1 signaling.
Collapse
|
33
|
Yetnikoff L, Pokinko M, Arvanitogiannis A, Flores C. Adolescence: a time of transition for the phenotype of dcc heterozygous mice. Psychopharmacology (Berl) 2014; 231:1705-14. [PMID: 23572211 DOI: 10.1007/s00213-013-3083-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/13/2013] [Indexed: 11/29/2022]
Abstract
RATIONALE Stark differences exist between adult (>PND 70) and juvenile (∼PND 21-34) rodents in how DCC (deleted in colorectal cancer) receptors and sensitization to amphetamine interact. In adults, repeated amphetamine upregulates DCC receptor expression selectively in the ventral tegmental area (VTA), an effect that is critical for sensitization. In contrast, amphetamine administered to juveniles downregulates VTA DCC expression. Moreover, whereas adult dcc heterozygous mice fail to sensitize when repeatedly treated with amphetamine, drug treatment during the juvenile period actually abolishes this adult "protective" phenotype. OBJECTIVES We set out to determine whether adolescence (PND ∼35-55) is a period during which: (1) amphetamine-induced alterations in VTA DCC expression switch from downregulation to upregulation; (2) the "protective" phenotype of adult dcc heterozygotes against sensitization becomes evident; and (3) the adult "protective" phenotype of dcc heterozygotes can still be abolished by repeated amphetamine treatment. RESULTS Repeated amphetamine did not significantly alter VTA DCC expression in adolescent rodents when assessed 1 week later. Both wild-type and dcc heterozygous mice exhibited sensitization at this time. Remarkably, wild-type mice, but not dcc heterozygotes, exhibited sensitization when tested during adulthood. CONCLUSIONS Adolescence is a time of transition for dcc heterozygotes as related to sensitization. Our results support the hypothesis that DCC may be a key factor in determining age-dependent individual differences in vulnerability to sensitization. Given that exposure to drugs of abuse during adolescence can have profound consequences for adulthood, the resilience of adult dcc heterozygous mice against adolescent exposure to amphetamine is particularly salient.
Collapse
Affiliation(s)
- Leora Yetnikoff
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | | | | | | |
Collapse
|
34
|
Hegarty SV, Collins LM, Gavin AM, Roche SL, Wyatt SL, Sullivan AM, O'Keeffe GW. Canonical BMP-Smad signalling promotes neurite growth in rat midbrain dopaminergic neurons. Neuromolecular Med 2014; 16:473-89. [PMID: 24682653 DOI: 10.1007/s12017-014-8299-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/07/2014] [Indexed: 01/01/2023]
Abstract
Ventral midbrain (VM) dopaminergic (DA) neurons project to the dorsal striatum via the nigrostriatal pathway to regulate voluntary movements, and their loss leads to the motor dysfunction seen in Parkinson's disease (PD). Despite recent progress in the understanding of VM DA neurogenesis, the factors regulating nigrostriatal pathway development remain largely unknown. The bone morphogenetic protein (BMP) family regulates neurite growth in the developing nervous system and may contribute to nigrostriatal pathway development. Two related members of this family, BMP2 and growth differentiation factor (GDF)5, have neurotrophic effects, including promotion of neurite growth, on cultured VM DA neurons. However, the molecular mechanisms regulating their effects on DA neurons are unknown. By characterising the temporal expression profiles of endogenous BMP receptors (BMPRs) in the developing and adult rat VM and striatum, this study identified BMP2 and GDF5 as potential regulators of nigrostriatal pathway development. Furthermore, through the use of noggin, dorsomorphin and BMPR/Smad plasmids, this study demonstrated that GDF5- and BMP2-induced neurite outgrowth from cultured VM DA neurons is dependent on BMP type I receptor activation of the Smad 1/5/8 signalling pathway.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| | | | | | | | | | | | | |
Collapse
|
35
|
Maintaining and modifying connections: roles for axon guidance cues in the mature nervous system. Neuropsychopharmacology 2014; 39:246-7. [PMID: 24317322 PMCID: PMC3857654 DOI: 10.1038/npp.2013.212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
36
|
Daubaras M, Dal Bo G, Flores C. Target-dependent expression of the netrin-1 receptor, UNC5C, in projection neurons of the ventral tegmental area. Neuroscience 2013; 260:36-46. [PMID: 24333968 DOI: 10.1016/j.neuroscience.2013.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/02/2013] [Accepted: 12/03/2013] [Indexed: 12/22/2022]
Abstract
We have shown previously that the netrin-1 receptor, unc-5 homologue C (UNC5C), is expressed by ventral tegmental area (VTA) dopamine (DA) neurons of rodents, but only from adolescence onwards (Manitt et al., 2010; Auger et al., 2013). The goal of this study was to characterize the expression of UNC5C by these neurons. Specifically, we assessed whether UNC5C expression is selective to DA neurons that project to the medial prefrontal cortex (mPFC), which undergo significant maturation during the adolescent period. To this end, we injected fluorescent retrograde tracer beads into the mPFC, nucleus accumbens (NAcc) core, or NAcc lateral shell of adult male wild-type C57Bl/6J mice and processed their brains for tyrosine hydroxylase (TH) and UNC5C immunofluorescence 2-3weeks later. VTA neurons with any combination of these immunolabels were visualized and counted using optical fractionator stereology. Our analysis revealed two main findings: (1) there are no differences in the proportions of UNC5C-positive DA neurons projecting to the mPFC, NAcc core, or NAcc lateral shell, and (2) the proportion of non-DA UNC5C-positive neurons targeting the mPFC is greater than the proportions of non-DA UNC5C-positive neurons targeting the NAcc core or lateral shell. These findings show that, contrary to our hypothesis, DA neurons projecting to the mPFC do not express UNC5C selectively. However, UNC5C expression by non-DA VTA neurons is predominantly found in those projecting to the mPFC and, as such, may play a role in their function.
Collapse
Affiliation(s)
- M Daubaras
- Department of Psychiatry and Integrated Program in Neuroscience, McGill University, Douglas Hospital Research Centre, 6875 LaSalle Boulevard, Montreal, Quebec H4H 1R3, Canada
| | - G Dal Bo
- Department of Psychiatry and Integrated Program in Neuroscience, McGill University, Douglas Hospital Research Centre, 6875 LaSalle Boulevard, Montreal, Quebec H4H 1R3, Canada
| | - C Flores
- Department of Psychiatry and Integrated Program in Neuroscience, McGill University, Douglas Hospital Research Centre, 6875 LaSalle Boulevard, Montreal, Quebec H4H 1R3, Canada.
| |
Collapse
|
37
|
dcc orchestrates the development of the prefrontal cortex during adolescence and is altered in psychiatric patients. Transl Psychiatry 2013; 3:e338. [PMID: 24346136 PMCID: PMC4030324 DOI: 10.1038/tp.2013.105] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 10/21/2013] [Indexed: 01/19/2023] Open
Abstract
Adolescence is a period of heightened susceptibility to psychiatric disorders of medial prefrontal cortex (mPFC) dysfunction and cognitive impairment. mPFC dopamine (DA) projections reach maturity only in early adulthood, when their control over cognition becomes fully functional. The mechanisms governing this protracted and unique development are unknown. Here we identify dcc as the first DA neuron gene to regulate mPFC connectivity during adolescence and dissect the mechanisms involved. Reduction or loss of dcc from DA neurons by Cre-lox recombination increased mPFC DA innervation. Underlying this was the presence of ectopic DA fibers that normally innervate non-cortical targets. Altered DA input changed the anatomy and electrophysiology of mPFC circuits, leading to enhanced cognitive flexibility. All phenotypes only emerged in adulthood. Using viral Cre, we demonstrated that dcc organizes mPFC wiring specifically during adolescence. Variations in DCC may determine differential predisposition to mPFC disorders in humans. Indeed, DCC expression is elevated in brains of antidepressant-free subjects who committed suicide.
Collapse
|
38
|
Kim JH, Lavan D, Chen N, Flores C, Cooper H, Lawrence AJ. Netrin-1 receptor-deficient mice show age-specific impairment in drug-induced locomotor hyperactivity but still self-administer methamphetamine. Psychopharmacology (Berl) 2013; 230:607-16. [PMID: 23820928 DOI: 10.1007/s00213-013-3187-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 06/11/2013] [Indexed: 12/27/2022]
Abstract
RATIONALE The mesocorticolimbic dopamine system undergoes significant reorganization of neuronal connectivity and functional refinement during adolescence. Deleted in colorectal cancer (DCC), a receptor for the guidance cue netrin-1, is involved in this reorganization. Previous studies have shown that adult mice with a heterozygous (het) loss-of-function mutation in DCC exhibit impairments in sensitization and conditioned place preference (CPP) to psychostimulants. However, the commonly abused psychostimulant methamphetamine (METH) has not been assessed, and the role of DCC in drug self-administration remains to be established. OBJECTIVES Using dcc het mice and wildtype (WT) littermates, we extended previous findings on dcc haplodeficiency by examining self-administration of METH in adult mice, including cue-induced drug seeking following abstinence. We also examined hyperactivity, sensitization, and CPP to a METH-paired context in adult and adolescent mice. RESULTS While adult dcc het mice expressed largely similar METH self-administration and cue-induced drug seeking as WT littermates, they failed to modulate responding according to dose of METH. Compared to WT, both adult and adolescent dcc het mice expressed impaired locomotor hyperactivity to acute METH but nevertheless showed comparable behavioral sensitization. Conditioned hyperactivity increased with age in WT but not in dcc het mice. CONCLUSIONS Impaired METH-induced hyperactivity and dose-related responding in adult dcc het mice suggest that reduced DCC alters METH-related behaviors. Adolescence is identified as a vulnerable period during which impairment in hyperactivity due to reduced DCC can be overcome with repeated METH injections. Nevertheless, DCC appears to have a somewhat limited role in METH-consumption and seeking following abstinence.
Collapse
Affiliation(s)
- Jee Hyun Kim
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3010, VIC, Australia,
| | | | | | | | | | | |
Collapse
|
39
|
Auger ML, Schmidt ERE, Manitt C, Dal-Bo G, Pasterkamp RJ, Flores C. unc5c haploinsufficient phenotype: striking similarities with the dcc haploinsufficiency model. Eur J Neurosci 2013; 38:2853-63. [PMID: 23738838 DOI: 10.1111/ejn.12270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 04/08/2013] [Accepted: 05/03/2013] [Indexed: 01/28/2023]
Abstract
DCC and UNC5 homologs (UNC5H) are guidance cue receptors highly expressed by mesocorticolimbic dopamine neurons. We have shown that dcc heterozygous mice exhibit increased dopamine, but not norepinephrine, innervation and function in medial prefrontal cortex. Concomitantly, dcc heterozygotes show blunted mesolimbic dopamine release and behavioral responses to stimulant drugs. These changes appear only in adulthood. Recently, we found an adolescent emergence of UNC5H expression by dopamine neurons and co-expression of DCC and UNC5H by single dopamine cells. Here, we demonstrate selective expression of unc5 homolog c mRNA by dopamine neurons in adulthood. We show that unc5c haploinsufficiency results in diminished amphetamine-induced locomotion in male and female mice. This phenotype is identical to that produced by dcc haploinsufficiency and is observed after adolescence. Notably, and similar to dcc haploinsufficiency, unc5c haploinsufficiency leads to dramatic increases in tyrosine hydroxylase expression in the medial prefrontal cortex, but not in the nucleus accumbens. In contrast, medial prefrontal cortex dopamine-β-hydroxylase expression is not altered. We confirmed that UNC5C protein is reduced in the ventral tegmental area of unc5c heterozygous mice, but that DCC expression in this region remains unchanged. UNC5C receptors may also play a role in dopamine function and influence sensitivity to behavioral effects of stimulant drugs of abuse, at least upon first exposure. The striking similarities between the dcc and the unc5c haploinsufficient phenotypes raise the possibility that functions mediated by DCC/UNC5C complexes may be at play.
Collapse
Affiliation(s)
- Meagan L Auger
- Department of Psychiatry and of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, 6875 LaSalle Boulevard, Verdun, QC, Canada, H4H 1R3
| | | | | | | | | | | |
Collapse
|
40
|
Midbrain dopaminergic neurons: a review of the molecular circuitry that regulates their development. Dev Biol 2013; 379:123-38. [PMID: 23603197 DOI: 10.1016/j.ydbio.2013.04.014] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/27/2013] [Accepted: 04/12/2013] [Indexed: 12/21/2022]
Abstract
Dopaminergic (DA) neurons of the ventral midbrain (VM) play vital roles in the regulation of voluntary movement, emotion and reward. They are divided into the A8, A9 and A10 subgroups. The development of the A9 group of DA neurons is an area of intense investigation to aid the generation of these neurons from stem cell sources for cell transplantation approaches to Parkinson's disease (PD). This review discusses the molecular processes that are involved in the identity, specification, maturation, target innervation and survival of VM DA neurons during development. The complex molecular interactions of a number of genetic pathways are outlined, as well as recent advances in the mechanisms that regulate subset identity within the VM DA neuronal pool. A thorough understanding of the cellular and molecular mechanisms involved in the development of VM DA neurons will greatly facilitate the use of cell replacement therapy for the treatment of PD.
Collapse
|
41
|
Mihali A, Subramani S, Kaunitz G, Rayport S, Gaisler-Salomon I. Modeling resilience to schizophrenia in genetically modified mice: a novel approach to drug discovery. Expert Rev Neurother 2012; 12:785-99. [PMID: 22853787 DOI: 10.1586/ern.12.60] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Complex psychiatric disorders, such as schizophrenia, arise from a combination of genetic, developmental, environmental and social factors. These vulnerabilities can be mitigated by adaptive factors in each of these domains engendering resilience. Modeling resilience in mice using transgenic approaches offers a direct path to intervention, as resilience mutations point directly to therapeutic targets. As prototypes for this approach, we discuss the three mouse models of schizophrenia resilience, all based on modulating glutamatergic synaptic transmission. This motivates the broader development of schizophrenia resilience mouse models independent of specific pathophysiological hypotheses as a strategy for drug discovery. Three guiding validation criteria are presented. A resilience-oriented approach should identify pharmacologically tractable targets and in turn offer new insights into pathophysiological mechanisms.
Collapse
Affiliation(s)
- Andra Mihali
- Department of Psychiatry, Columbia University, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 62, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
42
|
Aguilar-Valles A, Jung S, Poole S, Flores C, Luheshi GN. Leptin and interleukin-6 alter the function of mesolimbic dopamine neurons in a rodent model of prenatal inflammation. Psychoneuroendocrinology 2012; 37:956-69. [PMID: 22133515 DOI: 10.1016/j.psyneuen.2011.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 10/07/2011] [Accepted: 11/04/2011] [Indexed: 12/21/2022]
Abstract
Maternal inflammation during critical stages of gestation is thought to underlie the link between prenatal infection and several neurodevelopmental psychiatric disorders in the offspring, including schizophrenia. Increased activity of mesolimbic dopamine (DA) neurons, a hallmark of psychosis, is found in offspring of rodents exposed to a prenatal inflammatory challenge but it is unclear how this effect is elicited. Using an experimental model of localized aseptic inflammation with turpentine oil (TURP) we sought to establish whether circulating interleukin-6 (IL-6) and leptin play a role in the effects of prenatal inflammation on DA neurons. Both mediators are involved in the systemic inflammatory response to immunogens, with IL-6 mediating the early phase, followed by leptin in the late phase of the response. Maternal treatment with TURP at gestational day (GD) 15 enhanced the locomotor response to the DA indirect agonist, amphetamine (AMPH), increased the expression of tyrosine hydroxylase (TH), an enzyme involved in DA synthesis, DA levels and the expression of the post-synaptic protein spinophilin in the nucleus accumbens (NAcc) in the adult offspring. All of these alterations were totally abolished by co-treating the pregnant dams with a neutralizing IL-6 antiserum. Neutralization of maternal leptin prevented the enhanced behavioral sensitization and elevation of DA and spinophilin in the NAcc but spared other changes regulated by IL-6, such as increased NAcc TH levels and acute locomotor response to AMPH. Our results provide novel evidence to suggest that prenatal surges in both maternal circulating IL-6 and leptin contribute to the appearance of sensitized DA function in the adult offspring.
Collapse
Affiliation(s)
- Argel Aguilar-Valles
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, 6875 LaSalle Boulevard, Montreal, Quebec, Canada.
| | | | | | | | | |
Collapse
|
43
|
Grant A, Fathalli F, Rouleau G, Joober R, Flores C. Association between schizophrenia and genetic variation in DCC: a case-control study. Schizophr Res 2012; 137:26-31. [PMID: 22418395 DOI: 10.1016/j.schres.2012.02.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 02/18/2012] [Accepted: 02/21/2012] [Indexed: 12/27/2022]
Abstract
Schizophrenia is a highly heritable neurodevelopmental disorder associated with alterations in synaptic connectivity. Deleted in colorectal cancer (DCC), a receptor for the guidance cue netrin-1, plays a pivotal role in organizing neuronal circuitry by guiding growing axons and dendrites to their correct targets and by influencing synaptic connectivity. Results from experiments we previously conducted in dcc-heterozygous mice show that DCC plays a critical role in the developmental organization of the mesocorticolimbic dopamine (DA) circuitry. Furthermore we have shown that reduced expression of DCC during development and/or throughout life confers resilience to the development of schizophrenia-like DA and behavioural abnormalities. Importantly, this "protective" phenotype only emerges after puberty. Here we assess whether DCC may contribute to the risk of schizophrenia. We examined single nucleotide polymorphisms (SNPs) located in the DCC gene for association with schizophrenia using a case-control sample consisting of 556 unrelated schizophrenic patients and 208 healthy controls. We found one SNP, rs2270954, to be nominally associated with schizophrenia; patients were less likely to be heterozygous at this locus and more likely to be homozygous for the minor allele (χ(2)=9.84, df=2, nominal p=0.0071). Intriguingly, this SNP is located within the 3' untranslated region, an area known to contain a number of regulatory sequences that determine the stability and translation efficacy of mRNA. These results, together with our previous findings from studies in rodents, point at DCC as a promising novel candidate gene that may contribute to the genetic basis behind individual differences in susceptibility to schizophrenia.
Collapse
Affiliation(s)
- Alanna Grant
- Integrated Program in Neuroscience, McGill University, Douglas Mental Health University Institute, 6875 LaSalle Boulevard, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
44
|
Juvenile exposure to methylphenidate reduces cocaine reward and alters netrin-1 receptor expression in adulthood. Behav Brain Res 2012; 229:202-7. [PMID: 22249134 DOI: 10.1016/j.bbr.2012.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 12/21/2011] [Accepted: 01/04/2012] [Indexed: 11/21/2022]
Abstract
The guidance cue netrin-1 acting on mesocorticolimbic dopamine (DA) neurons through its receptor DCC (deleted in colorectal cancer) has been implicated in the neuronal plasticity induced by psychostimulant drugs. We examined in C57/BL6 mice the effects of repeated juvenile methylphenidate (MPH) exposure on cocaine-reward sensitivity in adulthood and determined whether early MPH treatment alters adult expression of DCC in mesocorticolimbic DA regions. By using place conditioning, we show that adult mice exposed to MPH during the juvenile period are less sensitive to cocaine-reward compared to saline-controls. Furthermore, by means of immunoblotting, we demonstrate that early MPH treatment attenuates adult DCC expression in the ventral tegmental area (VTA) selectively. These results support previous evidence that developmental MPH treatment diminishes cocaine-reward in adulthood and are the first to suggest that DCC in the VTA may participate in this enduring effect.
Collapse
|
45
|
Abolition of the behavioral phenotype of adult netrin-1 receptor deficient mice by exposure to amphetamine during the juvenile period. Psychopharmacology (Berl) 2011; 217:505-14. [PMID: 21523346 DOI: 10.1007/s00213-011-2312-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 04/07/2011] [Indexed: 10/18/2022]
Abstract
RATIONALE Netrin-1 guidance cues contribute to amphetamine-induced plasticity of the adult mesocorticolimbic dopamine system in rodents. The netrin-1 receptor, deleted in colorectal cancer (DCC), is upregulated by repeated amphetamine treatment selectively in the ventral tegmental area (VTA) of adult rats and wild-type mice. Furthermore, adult dcc heterozygous mice fail to show amphetamine-induced increases in VTA DCC expression and do not develop sensitization to this drug. OBJECTIVES The effects of netrin-1 receptor signaling on mesocorticolimbic dopamine system function change across development. However, the effects of AMPH on DCC receptor regulation and behavioral sensitization before puberty have not been determined. Here we examined whether (1) repeated amphetamine treatment would also alter DCC expression in juvenile rats and wild-type mice, and (2) dcc heterozygotes treated with amphetamine during the juvenile period (PND 22-32) would develop behavioral sensitization to this drug. RESULTS Repeated amphetamine downregulates DCC expression selectively in the VTA of juvenile rodents. Moreover, the behavioral phenotype of adult dcc heterozygous mice is not present before puberty and is abolished by amphetamine treatment during the juvenile period. Remarkably, adult dcc heterozygotes pretreated with amphetamine as juveniles no longer exhibit reduced DCC expression in the VTA compared to wild-type controls. CONCLUSIONS Our results indicate that netrin-1 receptor signaling may be a key factor in determining individual differences in vulnerability to the behaviorally sensitizing effects of amphetamine at different ages. Moreover, they suggest that the juvenile period marks a window of vulnerability during which exposure to stimulant drugs can reverse the behavioral phenotype of adult dcc heterozygous mice.
Collapse
|
46
|
Autocrine netrin function inhibits glioma cell motility and promotes focal adhesion formation. PLoS One 2011; 6:e25408. [PMID: 21980448 PMCID: PMC3182204 DOI: 10.1371/journal.pone.0025408] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 09/02/2011] [Indexed: 01/09/2023] Open
Abstract
Deregulation of mechanisms that control cell motility plays a key role in tumor progression by promoting tumor cell dissemination. Secreted netrins and their receptors, Deleted in Colorectal Cancer (DCC), neogenin, and the UNC5 homologues, regulate cell and axon migration, cell adhesion, and tissue morphogenesis. Netrin and netrin receptor expression have previously been shown to be disrupted in invasive tumors, including glioblastoma. We determined that the human glioblastoma cell lines U87, U343, and U373 all express neogenin, UNC5 homologues, and netrin-1 or netrin-3, but only U87 cells express DCC. Using transfilter migration assays, we demonstrate DCC-dependent chemoattractant migration of U87 cells up a gradient of netrin-1. In contrast, U343 and U373 cells, which do not express DCC, were neither attracted nor repelled. Ectopic expression of DCC by U343 and U373 cells resulted in these cells becoming competent to respond to a gradient of netrin-1 as a chemoattractant, and also slowed their rate of spontaneous migration. Here, in addition to netrins' well-characterized chemotropic activity, we demonstrate an autocrine function for netrin-1 and netrin-3 in U87 and U373 cells that slows migration. We provide evidence that netrins promote the maturation of focal complexes, structures associated with cell movement, into focal adhesions. Consistent with this, netrin, DCC, and UNC5 homologues were associated with focal adhesions, but not focal complexes. Disrupting netrin or DCC function did not alter cell proliferation or survival. Our findings provide evidence that DCC can slow cell migration, and that neogenin and UNC5 homologues are not sufficient to substitute for DCC function in these cells. Furthermore, we identify a role for netrins as autocrine inhibitors of cell motility that promote focal adhesion formation. These findings suggest that disruption of netrin signalling may disable a mechanism that normally restrains inappropriate cell migration.
Collapse
|
47
|
Flores C. Role of netrin-1 in the organization and function of the mesocorticolimbic dopamine system. J Psychiatry Neurosci 2011; 36:296-310. [PMID: 21481303 PMCID: PMC3163646 DOI: 10.1503/jpn.100171] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Changes in mesocorticolimbic dopamine (DA) neurons and their target cells can be induced throughout life and are important determinants of individual differences in susceptibility to psychopathology. The goal of my research is to gain insight into the nature of the cellularand molecular mechanism underlying the selective plasticity of mesocorticolimbic DA neurons. Here, I review work showing that the guidance cue netrin-1 is implicated in the organization, plasticity and function of mesocorticolimbic DA neurons in rodents. Developmental variations in netrin-1 receptor function result in selective reorganization of medial prefrontal DA circuitry during adolescence and in an adult phenotype protected against schizophrenia-like dopaminergic and behavioural abnormalities. Furthermore, in adulthood, expression of netrin-1 receptors is upregulated by repeated exposure to stimulant drugs of abuse in DA somatodendritic regions and is necessary for drug-induced behavioural plasticity. I propose that risk factors associated with DA-related adult psychiatric disorders alter netrin-1 function.
Collapse
Affiliation(s)
- Cecilia Flores
- Department of Psychiatry, McGill University, Douglas Hospital Research Centre, Montréal, QC, Canada.
| |
Collapse
|
48
|
The netrin receptor DCC is required in the pubertal organization of mesocortical dopamine circuitry. J Neurosci 2011; 31:8381-94. [PMID: 21653843 DOI: 10.1523/jneurosci.0606-11.2011] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Netrins are guidance cues involved in neural connectivity. We have shown that the netrin-1 receptor DCC (deleted in colorectal cancer) is involved in the functional organization of the mesocorticolimbic dopamine (DA) system. Adult mice with a heterozygous loss-of-function mutation in dcc exhibit changes in indexes of DA function, including DA-related behaviors. These phenotypes are only observed after puberty, a critical period in the maturation of the mesocortical DA projection. Here, we examined whether dcc heterozygous mice exhibit structural changes in medial prefrontal cortex (mPFC) DA synaptic connectivity, before and after puberty. Stereological counts of tyrosine-hydroxylase (TH)-positive varicosities were increased in the cingulate 1 and prelimbic regions of the pregenual mPFC. dcc heterozygous mice also exhibited alterations in the size, complexity, and dendritic spine density of mPFC layer V pyramidal neuron basilar dendritic arbors. Remarkably, these presynaptic and postsynaptic partner phenotypes were not observed in juvenile mice, suggesting that DCC selectively influences the extensive branching and synaptic differentiation that occurs in the maturing mPFC DA circuit at puberty. Immunolabeling experiments in wild-type mice demonstrated that DCC is segregated to TH-positive fibers innervating the nucleus accumbens, with only scarce DCC labeling in mPFC TH-positive fibers. Netrin had an inverted target expression pattern. Thus, DCC-mediated netrin-1 signaling may influence the formation/maintenance of mesocorticolimbic DA topography. In support of this, we report that dcc heterozygous mice exhibit a twofold increase in the density of mPFC DCC/TH-positive varicosities. Our results implicate DCC-mediated netrin-1 signaling in the establishment of mPFC DA circuitry during puberty.
Collapse
|
49
|
Lai Wing Sun K, Correia JP, Kennedy TE. Netrins: versatile extracellular cues with diverse functions. Development 2011; 138:2153-69. [PMID: 21558366 DOI: 10.1242/dev.044529] [Citation(s) in RCA: 318] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Netrins are secreted proteins that were first identified as guidance cues, directing cell and axon migration during neural development. Subsequent findings have demonstrated that netrins can influence the formation of multiple tissues, including the vasculature, lung, pancreas, muscle and mammary gland, by mediating cell migration, cell-cell interactions and cell-extracellular matrix adhesion. Recent evidence also implicates the ongoing expression of netrins and netrin receptors in the maintenance of cell-cell organisation in mature tissues. Here, we review the mechanisms involved in netrin signalling in vertebrate and invertebrate systems and discuss the functions of netrin signalling during the development of neural and non-neural tissues.
Collapse
Affiliation(s)
- Karen Lai Wing Sun
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | | | | |
Collapse
|
50
|
Dugan JP, Stratton A, Riley HP, Farmer WT, Mastick GS. Midbrain dopaminergic axons are guided longitudinally through the diencephalon by Slit/Robo signals. Mol Cell Neurosci 2011; 46:347-56. [PMID: 21118670 PMCID: PMC3021181 DOI: 10.1016/j.mcn.2010.11.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 11/01/2010] [Accepted: 11/15/2010] [Indexed: 01/21/2023] Open
Abstract
Dopaminergic neurons from the ventral mesencephalon/diencephalon (mesodiencephalon) form vital pathways constituting the majority of the brain's dopamine systems. Mesodiencephalic dopaminergic (mdDA) neurons extend longitudinal projections anteriorly through the diencephalon, ascending toward forebrain targets. The mechanisms by which mdDA axons initially navigate through the diencephalon are poorly understood. Recently the Slit family of secreted axon guidance proteins, and their Robo receptors, have been identified as important guides for descending longitudinal axons. To test the potential roles of Slit/Robo guidance in ascending trajectories, we examined tyrosine hydroxylase-positive (TH+) projections from mdDA neurons in mutant mouse embryos. We found that mdDA axons grow out of and parallel to Slit-positive ventral regions within the diencephalon, and that subsets of the mdDA axons likely express Robo1 and possibly also Robo2. Slit2 was able to directly inhibit TH axon outgrowth in explant co-culture assays. The mdDA axons made significant pathfinding errors in Slit1/2 and Robo1/2 knockout mice, including spreading out in the diencephalon to form a wider tract. The wider tract resulted from a combination of invasion of the ventral midline, consistent with Slit repulsion, but also axons wandering dorsally, away from the ventral midline. Aberrant dorsal trajectories were prominent in Robo1 and Robo1/2 knockout mice, suggesting that an aspect of Robo receptor function is Slit-independent. These results indicate that Slit/Robo signaling is critical during the initial establishment of dopaminergic pathways, with roles in the dorsoventral positioning and precise pathfinding of these ascending longitudinal axons.
Collapse
Affiliation(s)
- James P Dugan
- Department of Biology, University of Nevada, Reno, NV 89557
| | | | - Hilary P Riley
- Department of Biology, University of Nevada, Reno, NV 89557
| | - W Todd Farmer
- Department of Biology, University of Nevada, Reno, NV 89557
| | | |
Collapse
|