1
|
Broomer MC, Beacher NJ, Wang MW, Lin DT. Examining a punishment-related brain circuit with miniature fluorescence microscopes and deep learning. ADDICTION NEUROSCIENCE 2024; 11:100154. [PMID: 38680653 PMCID: PMC11044849 DOI: 10.1016/j.addicn.2024.100154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
In humans experiencing substance use disorder (SUD), abstinence from drug use is often motivated by a desire to avoid some undesirable consequence of further use: health effects, legal ramifications, etc. This process can be experimentally modeled in rodents by training and subsequently punishing an operant response in a context-induced reinstatement procedure. Understanding the biobehavioral mechanisms underlying punishment learning is critical to understanding both abstinence and relapse in individuals with SUD. To date, most investigations into the neural mechanisms of context-induced reinstatement following punishment have utilized discrete loss-of-function manipulations that do not capture ongoing changes in neural circuitry related to punishment-induced behavior change. Here, we describe a two-pronged approach to analyzing the biobehavioral mechanisms of punishment learning using miniature fluorescence microscopes and deep learning algorithms. We review recent advancements in both techniques and consider a target neural circuit.
Collapse
Affiliation(s)
- Matthew C. Broomer
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Nicholas J. Beacher
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Michael W. Wang
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Da-Ting Lin
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
2
|
Ayon-Olivas M, Wolf D, Andreska T, Granado N, Lüningschrör P, Ip CW, Moratalla R, Sendtner M. Dopaminergic Input Regulates the Sensitivity of Indirect Pathway Striatal Spiny Neurons to Brain-Derived Neurotrophic Factor. BIOLOGY 2023; 12:1360. [PMID: 37887070 PMCID: PMC10604681 DOI: 10.3390/biology12101360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Motor dysfunction in Parkinson's disease (PD) is closely linked to the dopaminergic depletion of striatal neurons and altered synaptic plasticity at corticostriatal synapses. Dopamine receptor D1 (DRD1) stimulation is a crucial step in the formation of long-term potentiation (LTP), whereas dopamine receptor D2 (DRD2) stimulation is needed for the formation of long-term depression (LTD) in striatal spiny projection neurons (SPNs). Tropomyosin receptor kinase B (TrkB) and its ligand brain-derived neurotrophic factor (BDNF) are centrally involved in plasticity regulation at the corticostriatal synapses. DRD1 activation enhances TrkB's sensitivity for BDNF in direct pathway spiny projection neurons (dSPNs). In this study, we showed that the activation of DRD2 in cultured striatal indirect pathway spiny projection neurons (iSPNs) and cholinergic interneurons causes the retraction of TrkB from the plasma membrane. This provides an explanation for the opposing synaptic plasticity changes observed upon DRD1 or DRD2 stimulation. In addition, TrkB was found within intracellular structures in dSPNs and iSPNs from Pitx3-/- mice, a genetic model of PD with early onset dopaminergic depletion in the dorsolateral striatum (DLS). This dysregulated BDNF/TrkB signaling might contribute to the pathophysiology of direct and indirect pathway striatal projection neurons in PD.
Collapse
Affiliation(s)
- Maurilyn Ayon-Olivas
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| | - Daniel Wolf
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| | - Thomas Andreska
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| | - Noelia Granado
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Patrick Lüningschrör
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
- CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| |
Collapse
|
3
|
Fung S, Smith CL, Prater KE, Case A, Green K, Osnis L, Winston C, Kinoshita Y, Sopher B, Morrison RS, Garden GA, Jayadev S. Early-Onset Familial Alzheimer Disease Variant PSEN2 N141I Heterozygosity is Associated with Altered Microglia Phenotype. J Alzheimers Dis 2020; 77:675-688. [PMID: 32741831 PMCID: PMC7592656 DOI: 10.3233/jad-200492] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Early-onset familial Alzheimer disease (EOFAD) is caused by heterozygous variants in the presenilin 1 (PSEN1), presenilin 2 (PSEN2), and APP genes. Decades after their discovery, the mechanisms by which these genes cause Alzheimer's disease (AD) or promote AD progression are not fully understood. While it is established that presenilin (PS) enzymatic activity produces amyloid-β (Aβ), PSs also regulate numerous other cellular functions, some of which intersect with known pathogenic drivers of neurodegeneration. Accumulating evidence suggests that microglia, resident innate immune cells in the central nervous system, play a key role in AD neurodegeneration. OBJECTIVE Previous work has identified a regulatory role for PS2 in microglia. We hypothesized that PSEN2 variants lead to dysregulated microglia, which could further contribute to disease acceleration. To mimic the genotype of EOFAD patients, we created a transgenic mouse expressing PSEN2 N141I on a mouse background expressing one wildtype PS2 and two PS1 alleles. RESULTS Microglial expression of PSEN2 N141I resulted in impaired γ-secretase activity as well as exaggerated inflammatory cytokine release, NFκB activity, and Aβ internalization. In vivo, PS2 N141I mice showed enhanced IL-6 and TREM2 expression in brain as well as reduced branch number and length, an indication of "activated" morphology, in the absence of inflammatory stimuli. LPS intraperitoneal injection resulted in higher inflammatory gene expression in PS2 N141I mouse brain relative to controls. CONCLUSION Our findings demonstrate that PSEN2 N141I heterozygosity is associated with disrupted innate immune homeostasis, suggesting EOFAD variants may promote disease progression through non-neuronal cells beyond canonical dysregulated Aβ production.
Collapse
Affiliation(s)
- Susan Fung
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Carole L. Smith
- Department of Neurology, University of Washington, Seattle, WA, USA
| | | | - Amanda Case
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Kevin Green
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Leah Osnis
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Chloe Winston
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Yoshito Kinoshita
- Department of Neurosurgery, University of Washington, Seattle, WA, USA
| | - Bryce Sopher
- Department of Neurology, University of Washington, Seattle, WA, USA
| | | | - Gwenn A. Garden
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA,Dr. Suman Jayadev, Department of Neurology, Box 356465, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA. Tel.: +1 206 221 2930;
| |
Collapse
|
4
|
Garr E, Delamater AR. Chemogenetic inhibition in the dorsal striatum reveals regional specificity of direct and indirect pathway control of action sequencing. Neurobiol Learn Mem 2020; 169:107169. [PMID: 31972244 DOI: 10.1016/j.nlm.2020.107169] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/07/2020] [Accepted: 01/18/2020] [Indexed: 11/17/2022]
Abstract
Animals engage in intricate action sequences that are constructed during instrumental learning. There is broad consensus that the basal ganglia play a crucial role in the formation and fluid performance of action sequences. To investigate the role of the basal ganglia direct and indirect pathways in action sequencing, we virally expressed Cre-dependent Gi-DREADDs in either the dorsomedial (DMS) or dorsolateral (DLS) striatum during and/or after action sequence learning in D1 and D2 Cre rats. Action sequence performance in D1 Cre rats was slowed down early in training when DREADDs were activated in the DMS, but sped up when activated in the DLS. Acquisition of the reinforced sequence was hindered when DREADDs were activated in the DLS of D2 Cre rats. Outcome devaluation tests conducted after training revealed that the goal-directed control of action sequence rates was immune to chemogenetic inhibition-rats suppressed the rate of sequence performance when rewards were devalued. Sequence initiation latencies were generally sensitive to outcome devaluation, except in the case where DREADD activation was removed in D2 Cre rats that previously experienced DREADD activation in the DMS during training. Sequence completion latencies were generally not sensitive to outcome devaluation, except in the case where D1 Cre rats experienced DREADD activation in the DMS during training and test. Collectively, these results suggest that the indirect pathway originating from the DLS is part of a circuit involved in the effective reinforcement of action sequences, while the direct and indirect pathways originating from the DMS contribute to the goal-directed control of sequence completion and initiation, respectively.
Collapse
Affiliation(s)
- Eric Garr
- Graduate Center, City University of New York, United States; Brooklyn College, City University of New York, United States.
| | - Andrew R Delamater
- Graduate Center, City University of New York, United States; Brooklyn College, City University of New York, United States
| |
Collapse
|
5
|
Garr E. Contributions of the basal ganglia to action sequence learning and performance. Neurosci Biobehav Rev 2019; 107:279-295. [PMID: 31541637 DOI: 10.1016/j.neubiorev.2019.09.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/22/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022]
Abstract
Animals engage in intricately woven and choreographed action sequences that are constructed from trial-and-error learning. The mechanisms by which the brain links together individual actions which are later recalled as fluid chains of behavior are not fully understood, but there is broad consensus that the basal ganglia play a crucial role in this process. This paper presents a comprehensive review of the role of the basal ganglia in action sequencing, with a focus on whether the computational framework of reinforcement learning can capture key behavioral features of sequencing and the neural mechanisms that underlie them. While a simple neurocomputational model of reinforcement learning can capture key features of action sequence learning, this model is not sufficient to capture goal-directed control of sequences or their hierarchical representation. The hierarchical structure of action sequences, in particular, poses a challenge for building better models of action sequencing, and it is in this regard that further investigations into basal ganglia information processing may be informative.
Collapse
Affiliation(s)
- Eric Garr
- Graduate Center, City University of New York, 365 5(th) Avenue, New York, NY 10016, United States.
| |
Collapse
|
6
|
Melzer S, Gil M, Koser DE, Michael M, Huang KW, Monyer H. Distinct Corticostriatal GABAergic Neurons Modulate Striatal Output Neurons and Motor Activity. Cell Rep 2018; 19:1045-1055. [PMID: 28467898 PMCID: PMC5437725 DOI: 10.1016/j.celrep.2017.04.024] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 03/21/2017] [Accepted: 04/09/2017] [Indexed: 01/04/2023] Open
Abstract
The motor cortico-basal ganglion loop is critical for motor planning, execution, and learning. Balanced excitation and inhibition in this loop is crucial for proper motor output. Excitatory neurons have been thought to be the only source of motor cortical input to the striatum. Here, we identify long-range projecting GABAergic neurons in the primary (M1) and secondary (M2) motor cortex that target the dorsal striatum. This population of projecting GABAergic neurons comprises both somatostatin-positive (SOM+) and parvalbumin-positive (PV+) neurons that target direct and indirect pathway striatal output neurons as well as cholinergic interneurons differentially. Notably, optogenetic stimulation of M1 PV+ and M2 SOM+ projecting neurons reduced locomotion, whereas stimulation of M1 SOM+ projecting neurons enhanced locomotion. Thus, corticostriatal GABAergic projections modulate striatal output and motor activity. Long-range GABAergic projections from the motor cortex directly innervate the striatum M1 and M2 long-range SOM+ and PV+ neurons differentially innervate striatal neurons Striatal cholinergic neurons are innervated mainly by M1 SOM+ projecting neurons Motor cortex PV+ and SOM+ projecting neurons differentially modulate locomotion
Collapse
Affiliation(s)
- Sarah Melzer
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Mariana Gil
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - David E Koser
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Magdalena Michael
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Kee Wui Huang
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Hannah Monyer
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
7
|
Moehle MS, Pancani T, Byun N, Yohn SE, Wilson GH, Dickerson JW, Remke DH, Xiang Z, Niswender CM, Wess J, Jones CK, Lindsley CW, Rook JM, Conn PJ. Cholinergic Projections to the Substantia Nigra Pars Reticulata Inhibit Dopamine Modulation of Basal Ganglia through the M 4 Muscarinic Receptor. Neuron 2017; 96:1358-1372.e4. [PMID: 29268098 PMCID: PMC5753765 DOI: 10.1016/j.neuron.2017.12.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 10/04/2017] [Accepted: 12/04/2017] [Indexed: 12/16/2022]
Abstract
Cholinergic regulation of dopaminergic inputs into the striatum is critical for normal basal ganglia (BG) function. This regulation of BG function is thought to be primarily mediated by acetylcholine released from cholinergic interneurons (ChIs) acting locally in the striatum. We now report a combination of pharmacological, electrophysiological, optogenetic, chemogenetic, and functional magnetic resonance imaging studies suggesting extra-striatal cholinergic projections from the pedunculopontine nucleus to the substantia nigra pars reticulata (SNr) act on muscarinic acetylcholine receptor subtype 4 (M4) to oppose cAMP-dependent dopamine receptor subtype 1 (D1) signaling in presynaptic terminals of direct pathway striatal spiny projections neurons. This induces a tonic inhibition of transmission at direct pathway synapses and D1-mediated activation of motor activity. These studies provide important new insights into the unique role of M4 in regulating BG function and challenge the prevailing hypothesis of the centrality of striatal ChIs in opposing dopamine regulation of BG output.
Collapse
Affiliation(s)
- Mark S Moehle
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Tristano Pancani
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Nellie Byun
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA
| | - Samantha E Yohn
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - George H Wilson
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA
| | - Johnathan W Dickerson
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Daniel H Remke
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Zixiu Xiang
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Colleen M Niswender
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - Carrie K Jones
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jerri M Rook
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery and Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
8
|
Abstract
The basal ganglia are a series of interconnected subcortical nuclei. The function and dysfunction of these nuclei have been studied intensively in motor control, but more recently our knowledge of these functions has broadened to include prominent roles in cognition and affective control. This review summarizes historical models of basal ganglia function, as well as findings supporting or conflicting with these models, while emphasizing recent work in animals and humans directly testing the hypotheses generated by these models.
Collapse
|
9
|
Jiang L, O'Leary C, Kim HA, Parish CL, Massalas J, Waddington JL, Ehrlich ME, Schütz G, Gantois I, Lawrence AJ, Drago J. Motor and behavioral phenotype in conditional mutants with targeted ablation of cortical D1 dopamine receptor-expressing cells. Neurobiol Dis 2015; 76:137-158. [PMID: 25684539 DOI: 10.1016/j.nbd.2015.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 01/14/2015] [Accepted: 02/05/2015] [Indexed: 10/24/2022] Open
Abstract
D1-dopamine receptors (Drd1a) are highly expressed in the deep layers of the cerebral cortex and the striatum. A number of human diseases such as Huntington disease and schizophrenia are known to have cortical pathology involving dopamine receptor expressing neurons. To illuminate their functional role, we exploited a Cre/Lox molecular paradigm to generate Emx-1(tox) MUT mice, a transgenic line in which cortical Drd1a-expressing pyramidal neurons were selectively ablated. Emx-1(tox) MUT mice displayed prominent forelimb dystonia, hyperkinesia, ataxia on rotarod testing, heightened anxiety-like behavior, and age-dependent abnormalities in a test of social interaction. The latter occurred in the context of normal working memory on testing in the Y-maze and for novel object recognition. Some motor and behavioral abnormalities in Emx-1(tox) MUT mice overlapped with those in CamKIIα(tox) MUT transgenic mice, a line in which both striatal and cortical Drd1a-expressing cells were ablated. Although Emx-1(tox) MUT mice had normal striatal anatomy, both Emx-1(tox) MUT and CamKIIα(tox) MUT mice displayed selective neuronal loss in cortical layers V and VI. This study shows that loss of cortical Drd1a-expressing cells is sufficient to produce deficits in multiple motor and behavioral domains, independent of striatal mechanisms. Primary cortical changes in the D1 dopamine receptor compartment are therefore likely to model a number of core clinical features in disorders such as Huntington disease and schizophrenia.
Collapse
Affiliation(s)
- Luning Jiang
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia; St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Claire O'Leary
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia; Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Hyun Ah Kim
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Clare L Parish
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Jim Massalas
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - John L Waddington
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Michelle E Ehrlich
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA
| | - Günter Schütz
- Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Ilse Gantois
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - John Drago
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia; St Vincent's Hospital, Melbourne, Victoria, Australia.
| |
Collapse
|
10
|
Urban DJ, Roth BL. DREADDs (designer receptors exclusively activated by designer drugs): chemogenetic tools with therapeutic utility. Annu Rev Pharmacol Toxicol 2014; 55:399-417. [PMID: 25292433 DOI: 10.1146/annurev-pharmtox-010814-124803] [Citation(s) in RCA: 462] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the past decade, emerging synthetic biology technologies such as chemogenetics have dramatically transformed how pharmacologists and systems biologists deconstruct the involvement of G protein-coupled receptors (GPCRs) in a myriad of physiological and translational settings. Here we highlight a specific chemogenetic application that extends the utility of the concept of RASSLs (receptors activated solely by synthetic ligands): We have dubbed it DREADDs (designer receptors exclusively activated by designer drugs). As we show in this review, DREADDs are now used ubiquitously to modulate GPCR activity noninvasively in vivo. Results from these studies have directly implicated GPCR signaling in a large number of therapeutically relevant contexts. We also highlight recent applications of DREADD technology that have illuminated GPCR signaling processes that control pathways relevant to the treatment of eating disorders, obesity, and obesity-associated metabolic abnormalities. Additionally, we provide an overview of the potential utility of chemogenetic technologies for transformative therapeutics.
Collapse
|
11
|
Kim HA, Jiang L, Madsen H, Parish CL, Massalas J, Smardencas A, O'Leary C, Gantois I, O'Tuathaigh C, Waddington JL, Ehrlich ME, Lawrence AJ, Drago J. Resolving pathobiological mechanisms relating to Huntington disease: gait, balance, and involuntary movements in mice with targeted ablation of striatal D1 dopamine receptor cells. Neurobiol Dis 2013; 62:323-37. [PMID: 24135007 DOI: 10.1016/j.nbd.2013.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 08/13/2013] [Accepted: 09/14/2013] [Indexed: 12/01/2022] Open
Abstract
Progressive cell loss is observed in the striatum, cerebral cortex, thalamus, hypothalamus, subthalamic nucleus and hippocampus in Huntington disease. In the striatum, dopamine-responsive medium spiny neurons are preferentially lost. Clinical features include involuntary movements, gait and orofacial impairments in addition to cognitive deficits and psychosis, anxiety and mood disorders. We utilized the Cre-LoxP system to generate mutant mice with selective postnatal ablation of D1 dopamine receptor-expressing striatal neurons to determine which elements of the complex Huntington disease phenotype relate to loss of this neuronal subpopulation. Mutant mice had reduced body weight, locomotor slowing, reduced rearing, ataxia, a short stride length wide-based erratic gait, impairment in orofacial movements and displayed haloperidol-suppressible tic-like movements. The mutation was associated with an anxiolytic profile. Mutant mice had significant striatal-specific atrophy and astrogliosis. D1-expressing cell number was reduced throughout the rostrocaudal extent of the dorsal striatum consistent with partial destruction of the striatonigral pathway. Additional striatal changes included up-regulated D2 and enkephalin mRNA, and an increased density of D2 and preproenkephalin-expressing projection neurons, and striatal neuropeptide Y and cholinergic interneurons. These data suggest that striatal D1-cell-ablation alone may account for the involuntary movements and locomotor, balance and orofacial deficits seen not only in HD but also in HD phenocopy syndromes with striatal atrophy. Therapeutic strategies would therefore need to target striatal D1 cells to ameliorate deficits especially when the clinical presentation is dominated by a bradykinetic/ataxic phenotype with involuntary movements.
Collapse
Affiliation(s)
- Hyun Ah Kim
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Luning Jiang
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Heather Madsen
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Clare L Parish
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Jim Massalas
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Arthur Smardencas
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Claire O'Leary
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia; Molecular and Cellular Therapeutics, RCSI Research Institute, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ilse Gantois
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Colm O'Tuathaigh
- Molecular and Cellular Therapeutics, RCSI Research Institute, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - John L Waddington
- Molecular and Cellular Therapeutics, RCSI Research Institute, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Michelle E Ehrlich
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - John Drago
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
12
|
Smardencas A, Rizkalla K, Kim HA, Massalas J, O'Leary C, Ehrlich ME, Schütz G, Lawrence AJ, Drago J. Phenotyping dividing cells in mouse models of neurodegenerative basal ganglia diseases. BMC Neurosci 2013; 14:111. [PMID: 24090101 PMCID: PMC3851877 DOI: 10.1186/1471-2202-14-111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 09/18/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mice generated by a Cre/LoxP transgenic paradigm were used to model neurodegenerative basal ganglia disease of which Huntington disease (HD) is the prototypical example. In HD, death occurs in striatal projection neurons as well as cortical neurons. Cortical and striatal neurons that express the D1 dopamine receptor (Drd1a) degenerate in HD. The contribution that death of specific neuronal cell populations makes to the HD disease phenotype and the response of the brain to loss of defined cell subtypes is largely unknown. METHODS Drd1a-expressing cells were targeted for cell death and three independent lines generated; a striatal-restricted line, a cortical-restricted line and a global line in which Drd1a cells were deleted from both the striatum and cortex. Two independent experimental approaches were used. In the first, the proliferative marker Ki-67 was used to identify proliferating cells in eighty-week-old mice belonging to a generic global line, a global in which Drd1a cells express green fluorescent protein (GFP-global) and in eighty-week-old mice of a cortical line. In the second experiment, the proliferative response of four-week-old mice belonging to GFP-global and striatal lines was assessed using the thymidine analogue BrdU. The phenotype of proliferating cells was ascertained by double staining for BrdU and Olig2 (an oligodendrocyte marker), Iba1 (a microglial cell marker), S100β (an astroglial cell marker), or NeuN (a neuronal cell marker). RESULTS In the first study, we found that Ki-67-expressing cells were restricted to the striatal side of the lateral ventricles. Control mice had a greater number of Ki-67+ cells than mutant mice. There was no overlap between Ki-67 and GFP staining in control or mutant mice, suggesting that cells did not undergo cell division once they acquired a Drd1a phenotype. In contrast, in the second study we found that BrdU+ cells were identified throughout the cortex, striatum and periventricular region of control and mutant mice. Mutant mice from the GFP-global line showed increased BrdU+ cells in the cortex, striatum and periventricular region relative to control. Striatal line mutant mice had an increased number of BrdU+ cells in the striatum and periventricular region, but not the cortex. The number of microglia, astrocytes, oligodendrocytes and neurons generated from dividing progenitors was increased relative to control mice in most brain regions in mutant mice from the GFP-global line. In contrast, striatal line mutant mice displayed an increase only in the number of dividing microglia in striatal and periventricular regions. CONCLUSIONS Genetically programmed post-natal ablation of Drd1a-expressing neurons is associated with an extensive proliferative response involving multiple cell lineages. The nature of the tissue response has the potential not only to remove cellular debris but also to forge physiologically meaningful brain repair. Age related deficits in proliferation are seen in mutant lines. A blunted endogenous reparative response may underlie the cumulative deficits characteristic of age related neurodegeneration.
Collapse
Affiliation(s)
- Arthur Smardencas
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Reiner A, Shelby E, Wang H, DeMarch Z, Deng Y, Guley NH, Hogg V, Roxburgh R, Tippett LJ, Waldvogel HJ, Faull RLM. Striatal parvalbuminergic neurons are lost in Huntington's disease: implications for dystonia. Mov Disord 2013; 28:1691-9. [PMID: 24014043 PMCID: PMC3812318 DOI: 10.1002/mds.25624] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 07/01/2013] [Accepted: 07/03/2013] [Indexed: 12/13/2022] Open
Abstract
Although dystonia represents a major source of motor disability in Huntington's disease (HD), its pathophysiology remains unknown. Because recent animal studies indicate that loss of parvalbuminergic (PARV+) striatal interneurons can cause dystonia, we investigated if loss of PARV+ striatal interneurons occurs during human HD progression, and thus might contribute to dystonia in HD. We used immunolabeling to detect PARV+ interneurons in fixed sections, and corrected for disease-related striatal atrophy by expressing PARV+ interneuron counts in ratio to interneurons co-containing somatostatin and neuropeptide Y (whose numbers are unaffected in HD). At all symptomatic HD grades, PARV+ interneurons were reduced to less than 26% of normal abundance in rostral caudate. In putamen rostral to the level of globus pallidus, loss of PARV+ interneurons was more gradual, not dropping off to less than 20% of control until grade 2. Loss of PARV+ interneurons was even more gradual in motor putamen at globus pallidus levels, with no loss at grade 1, and steady grade-wise decline thereafter. A large decrease in striatal PARV+ interneurons, thus, occurs in HD with advancing disease grade, with regional variation in the loss per grade. Given the findings of animal studies and the grade-wise loss of PARV+ striatal interneurons in motor striatum in parallel with the grade-wise appearance and worsening of dystonia, our results raise the possibility that loss of PARV+ striatal interneurons is a contributor to dystonia in HD.
Collapse
Affiliation(s)
- Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science CenterMemphis, Tennessee, USA
| | - Evan Shelby
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science CenterMemphis, Tennessee, USA
| | - Hongbing Wang
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science CenterMemphis, Tennessee, USA
| | - Zena DeMarch
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science CenterMemphis, Tennessee, USA
| | - Yunping Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science CenterMemphis, Tennessee, USA
| | - Natalie Hart Guley
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science CenterMemphis, Tennessee, USA
| | - Virginia Hogg
- Centre for Brain Research, University of AucklandAuckland, New Zealand
- Department of Psychology, University of AucklandAuckland, New Zealand
| | - Richard Roxburgh
- Centre for Brain Research, University of AucklandAuckland, New Zealand
- Department of Neurology, Auckland City HospitalAuckland, New Zealand
| | - Lynette J Tippett
- Centre for Brain Research, University of AucklandAuckland, New Zealand
- Department of Psychology, University of AucklandAuckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, University of AucklandAuckland, New Zealand
- Department of Anatomy with Radiology, University of AucklandAuckland, New Zealand
| | - Richard LM Faull
- Centre for Brain Research, University of AucklandAuckland, New Zealand
- Department of Anatomy with Radiology, University of AucklandAuckland, New Zealand
| |
Collapse
|
14
|
Abstract
Temporal control, or how organisms guide movements in time to achieve behavioral goals, depends on dopamine signaling. The medial prefrontal cortex controls many goal-directed behaviors and receives dopaminergic input primarily from the midbrain ventral tegmental area. However, this system has never been linked with temporal control. Here, we test the hypothesis that dopaminergic projections from the ventral tegmental area to the prefrontal cortex influence temporal control. Rodents were trained to perform a fixed-interval timing task with an interval of 20 s. We report several results: first, that decreasing dopaminergic neurotransmission using virally mediated RNA interference of tyrosine hydroxylase impaired temporal control, and second that pharmacological disruption of prefrontal D1 dopamine receptors, but not D2 dopamine receptors, impaired temporal control. We then used optogenetics to specifically and selectively manipulate prefrontal neurons expressing D1 dopamine receptors during fixed-interval timing performance. Selective inhibition of D1-expressing prefrontal neurons impaired fixed-interval timing, whereas stimulation made animals more efficient during task performance. These data provide evidence that ventral tegmental dopaminergic projections to the prefrontal cortex influence temporal control via D1 receptors. The results identify a critical circuit for temporal control of behavior that could serve as a target for the treatment of dopaminergic diseases.
Collapse
|
15
|
Fukabori R, Okada K, Nishizawa K, Kai N, Kobayashi K, Uchigashima M, Watanabe M, Tsutsui Y, Kobayashi K. Striatal direct pathway modulates response time in execution of visual discrimination. Eur J Neurosci 2012; 35:784-97. [DOI: 10.1111/j.1460-9568.2012.08005.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
16
|
Differential regulation of motor control and response to dopaminergic drugs by D1R and D2R neurons in distinct dorsal striatum subregions. EMBO J 2011; 31:640-53. [PMID: 22068054 DOI: 10.1038/emboj.2011.400] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/12/2011] [Indexed: 12/17/2022] Open
Abstract
The dorsal striatum is critically involved in a variety of motor behaviours, including regulation of motor activity, motor skill learning and motor response to psychostimulant and neuroleptic drugs, but contribution of D(2)R-striatopallidal and D(1)R-striatonigral neurons in the dorsomedial (DMS, associative) and dorsolateral (DLS, sensorimotor) striatum to distinct functions remains elusive. To delineate cell type-specific motor functions of the DMS or the DLS, we selectively ablated D(2)R- and D(1)R-expressing striatal neurons with spatial resolution. We found that associative striatum exerts a population-selective control over locomotion and reactivity to novelty, striatopallidal and striatonigral neurons inhibiting and stimulating exploration, respectively. Further, DMS-striatopallidal neurons are involved only in early motor learning whereas gradual motor skill acquisition depends on striatonigral neurons in the sensorimotor striatum. Finally, associative striatum D(2)R neurons are required for the cataleptic effect of the typical neuroleptic drug haloperidol and for amphetamine motor response sensitization. Altogether, these data provide direct experimental evidence for cell-specific topographic functional organization of the dorsal striatum.
Collapse
|
17
|
Kreitzer AC, Berke JD. Investigating striatal function through cell-type-specific manipulations. Neuroscience 2011; 198:19-26. [PMID: 21867745 DOI: 10.1016/j.neuroscience.2011.08.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/03/2011] [Accepted: 08/06/2011] [Indexed: 12/17/2022]
Abstract
The striatum integrates convergent input from the cortex, thalamus, and midbrain, and has a powerful influence over motivated behavior via outputs to downstream basal ganglia nuclei. Although the anatomy and physiology of distinct classes of striatal neurons have been intensively studied, the specific functions of these cell subpopulations have been more difficult to address. Recently, application of new methodologies for perturbing activity and signaling in different cell types in vivo has begun to allow direct tests of the causal roles of striatal neurons in behavior.
Collapse
Affiliation(s)
- A C Kreitzer
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA.
| | | |
Collapse
|
18
|
Tomiyama K, Kim HA, Kinsella A, Ehrlich ME, Schütz G, Koshikawa N, Lawrence AJ, Waddington JL, Drago J. Phenotypic disruption to orofacial movement topography in conditional mutants with generalized CamKIIa/Cre D1Tox versus striatal-specific DARPP-32/Cre D1Tox ablation of D1 dopamine receptor-expressing cells. Synapse 2011; 65:835-42. [PMID: 21308794 DOI: 10.1002/syn.20910] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 12/21/2010] [Indexed: 01/29/2023]
Abstract
Orofacial movements were quantified in (a) DARPP-32/Cre D1Tox mutants, having progressive loss of D1 dopamine receptor expressing striatal medium spiny neurons and (b) CamKIIa/Cre D1Tox mutants, having progressive, generalized loss of forebrain D1 receptor expressing cells. Horizontal jaw movements and tongue protrusions were reduced in DARPP-32/Cre but not in CamKIIa/Cre mutants; head and vibrissae movements were increased in DARPP-32/Cre but decreased in CamKIIa/Cre mutants. In drug challenge studies, tongue protrusions were increased in CamKIIa/Cre mutants following vehicle, suggesting a stress-related phenotype. These findings indicate that mice with progressive loss of striatal-specific D1 receptor expressing cells have an orofacial phenotype that may be modulated by the loss of extrastriatal D1 receptor expressing cells. As progressive loss of D1 dopamine receptor-expressing cells is a hallmark feature of Huntington's disease (HD), these findings may inform the functional role of loss of this cell population in the overall pathobiology of HD.
Collapse
Affiliation(s)
- Katsunori Tomiyama
- Advanced Research Institute for the Sciences and Humanities, Nihon University, Tokyo 102, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Fujimoto K, Araki K, McCarthy DM, Sims JR, Ren JQ, Zhang X, Bhide PG. A transgenic mouse model of neuroepithelial cell specific inducible overexpression of dopamine D1-receptor. Neuroscience 2010; 170:961-70. [PMID: 20674683 DOI: 10.1016/j.neuroscience.2010.07.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 07/14/2010] [Accepted: 07/18/2010] [Indexed: 01/01/2023]
Abstract
Dopamine and its receptors appear in the brain during early embryonic period suggesting a role for dopamine in brain development. In fact, dopamine receptor imbalance resulting from impaired physiological balance between D1- and D2-receptor activities can perturb brain development and lead to persisting changes in brain structure and function. Dopamine receptor imbalance can be produced experimentally using pharmacological or genetic methods. Pharmacological methods tend to activate or antagonize the receptors in all cell types. In the traditional gene knockout models the receptor imbalance occurs during development and also at maturity. Therefore, assaying the effects of dopamine imbalance on specific cell types (e.g. precursor versus postmitotic cells) or at specific periods of brain development (e.g. pre- or postnatal periods) is not feasible in these models. We describe a novel transgenic mouse model based on the tetracycline dependent inducible gene expression system in which dopamine D1-receptor transgene expression is induced selectively in neuroepithelial cells of the embryonic brain at experimenter-chosen intervals of brain development. In this model, doxycycline-induced expression of the transgene causes significant overexpression of the D1-receptor and significant reductions in the incorporation of the S-phase marker bromodeoxyuridine into neuroepithelial cells of the basal and dorsal telencephalon indicating marked effects on telencephalic neurogenesis. The D1-receptor overexpression occurs at higher levels in the medial ganglionic eminence (MGE) than the lateral ganglionic eminence (LGE) or cerebral wall (CW). Moreover, although the transgene is induced selectively in the neuroepithelium, D1-receptor protein overexpression appears to persist in postmitotic cells. The mouse model can be modified for neuroepithelial cell-specific inducible expression of other transgenes or induction of the D1-receptor transgene in other cells in specific brain regions by crossbreeding the mice with transgenic mouse lines available already.
Collapse
Affiliation(s)
- K Fujimoto
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Age-related behavioural phenotype and cellular characterisation of mice with progressive ablation of D1 dopamine receptor-expressing cells. Behav Brain Res 2009; 206:78-87. [PMID: 19733597 DOI: 10.1016/j.bbr.2009.08.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 08/25/2009] [Accepted: 08/29/2009] [Indexed: 11/23/2022]
Abstract
In this study we characterize the behavioural and cellular phenotype of mutant (MUT) mice with progressive loss of D1 dopamine receptor (Drd1a)-expressing cells. Adult [14-19 weeks] MUT mice showed intact working memory in the spontaneous alternation test but evidenced anxiety-like behaviour in the elevated plus maze and the light-dark test. The ethogram of mature adult MUT [average age 22 weeks] was compared with that of young adult MUT mice [average age 12 weeks]. While MUT mice evidenced hyperactivity over initial exploration at both time points, the topography of hyperactivity shifted. Moreover, initial hyperactivity was sustained over habituation at 12 weeks, but not at 22 weeks. Thus, by 22 weeks MUT mice evidenced shifts in, and mitigation of, these early phenotypic effects. However, orofacial behaviours of chewing and sifting were reduced similarly at 12 and 22 weeks. These data support the hypothesis that aspects of the mutant phenotype change with time. Quantitative autoradiography at 20 weeks revealed loss of D1-like dopamine receptor binding in the entire basal ganglia, with upregulated D2-like binding. There appear to be topographically specific interactions between normal maturational processes and compensatory mechanisms evoked subsequent to targeted ablation of D1 dopamine receptor-expressing cells. Understanding the mechanistic bases of mitigation vs persistence of individual phenotypes in relation to neural adaptation consequent to cell loss may lead to novel therapeutic strategies for basal ganglia disorders.
Collapse
|
21
|
Chen J, Rusnak M, Lombroso PJ, Sidhu A. Dopamine promotes striatal neuronal apoptotic death via ERK signaling cascades. Eur J Neurosci 2009; 29:287-306. [PMID: 19200235 DOI: 10.1111/j.1460-9568.2008.06590.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although the mechanisms underlying striatal neurodegeneration are poorly understood, we have shown that striatal pathogenesis may be initiated by high synaptic levels of extracellular dopamine (DA). Here we investigated in rat striatal primary neurons the mobilization of the mitogen-activated protein kinase (MAPK) signaling pathways after treatment with DA. Instead of observing an elevation of the archetypical pro-cytotoxic MAPKs, p-JNK and p-p38 MAPK, we found that DA, acting through D1 DA receptors, induced a sustained stimulation of the phosphorylated form of extracellular signal-regulated kinase (p-ERK) via a cAMP/protein kinase A (PKA)/Rap1/B-Raf / MAPK/ERK kinase (MEK) pathway. Blockade of D2 DA receptors, beta-adrenergic receptors or N-methyl-D-aspartate receptors with receptor-specific antagonists had no significant effect on this process. Activation of D1 DA receptors and PKA by DA caused phosphorylation and inactivation of the striatal-enriched tyrosine phosphatase, an important phosphatase for the dephosphorylation and subsequent inactivation of p-ERK in the striatum. Interestingly, p-ERK was primarily retained in the cytoplasm, with only low amounts translocated to the nucleus. The scaffold protein beta-arrestin2 interacted with both p-ERK and D1 DA receptor, triggering the cytosolic retention of p-ERK and inducing striatal neuronal apoptotic death. These data provide unique insight into a novel role of p-ERK in striatal neurodegeneration.
Collapse
Affiliation(s)
- Jun Chen
- Department of Pediatrics, Georgetown University, Washington DC 20007, USA
| | | | | | | |
Collapse
|
22
|
Kishioka A, Fukushima F, Ito T, Kataoka H, Mori H, Ikeda T, Itohara S, Sakimura K, Mishina M. A novel form of memory for auditory fear conditioning at a low-intensity unconditioned stimulus. PLoS One 2009; 4:e4157. [PMID: 19132103 PMCID: PMC2613534 DOI: 10.1371/journal.pone.0004157] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Accepted: 11/23/2008] [Indexed: 11/19/2022] Open
Abstract
Fear is one of the most potent emotional experiences and is an adaptive component of response to potentially threatening stimuli. On the other hand, too much or inappropriate fear accounts for many common psychiatric problems. Cumulative evidence suggests that the amygdala plays a central role in the acquisition, storage and expression of fear memory. Here, we developed an inducible striatal neuron ablation system in transgenic mice. The ablation of striatal neurons in the adult brain hardly affected the auditory fear learning under the standard condition in agreement with previous studies. When conditioned with a low-intensity unconditioned stimulus, however, the formation of long-term fear memory but not short-tem memory was impaired in striatal neuron-ablated mice. Consistently, the ablation of striatal neurons 24 h after conditioning with the low-intensity unconditioned stimulus, when the long-term fear memory was formed, diminished the retention of the long-term memory. Our results reveal a novel form of the auditory fear memory depending on striatal neurons at the low-intensity unconditioned stimulus.
Collapse
Affiliation(s)
- Ayumi Kishioka
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Fumiaki Fukushima
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tamae Ito
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hirotaka Kataoka
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hisashi Mori
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Toshio Ikeda
- Laboratory of Behavioral Genetics, Brain Science Institute, RIKEN, Saitama, Japan
| | - Shigeyoshi Itohara
- Laboratory of Behavioral Genetics, Brain Science Institute, RIKEN, Saitama, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masayoshi Mishina
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
23
|
Molecular profiling of striatonigral and striatopallidal medium spiny neurons past, present, and future. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 89:1-35. [PMID: 19900613 DOI: 10.1016/s0074-7742(09)89001-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Defining distinct molecular properties of the two striatal medium spiny neurons (MSNs) has been a challenging task for basal ganglia (BG) neuroscientists. Identifying differential molecular components in each MSN subtype is crucial for BG researchers to understand functional properties of these two neurons. The two MSN populations are morphologically identical except in their projections through the direct verses indirect BG pathways and they are heterogeneously dispersed throughout the dorsal striatum (dStr) and nucleus accumbens (NAc). These characteristics have made it difficult for researchers to distinguish and isolate these two neuronal populations thereby hindering progress toward a more comprehensive understanding of their differential molecular properties. Researchers began to investigate molecular differences in the striatonigral and striatopallidal neurons using in situ hybridization (ISH) techniques and single cell reverse transcription-polymerase chain reaction (scRT-PCR). Currently the field is utilizing more advanced techniques for large-scale gene expression studies including fluorescence activated cell sorting (FACS) of MSNs, from which RNA is purified, from fluorescent reporter transgenic mice or use of transgenic mice in which ribosomes from each MSN are tagged and can be immunoprecipitated followed by RNA isolation, a technique termed translating ribosomal affinity purification (TRAP). Additionally, the availability of fluorescent reporter mice for each MSN subtype is allowing, scientists to perform more accurate histology studies evaluating differential protein expression and signaling changes in each cell subtype. Finally, researchers are able to evaluate the role of specific genes in vivo by utilizing cell type-specific mouse models including Cre driver lines that can be crossed with conditional overexpression or knockout systems. This is a very exciting time in the BG field because researchers are well equipped with the most progressive tools to comprehensively evaluate molecular components in the two MSNs and their consequence on BG functional output in the normal, diseased, and developing brain.
Collapse
|
24
|
Heusner CL, Beutler LR, Houser CR, Palmiter RD. Deletion of GAD67 in dopamine receptor-1 expressing cells causes specific motor deficits. Genesis 2008; 46:357-67. [PMID: 18615733 PMCID: PMC3360952 DOI: 10.1002/dvg.20405] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The medium spiny neurons (MSNs), which comprise the direct and indirect output pathways from the striatum, use gamma-aminobutyric acid (GABA) as their major fact-acting neurotransmitter. We generated mice carrying a conditional allele of the Gad1 gene, which encodes GAD67, one of the two enzymes responsible for GABA biosynthesis, and bred them to mice expressing Cre recombinase at the dopamine D1 receptor locus (Drd1a) to selectively reduce GABA synthesis in the direct output pathway from the striatum. We show that these mice are deficient in some types of motor skills, but normal for others, suggesting a differential role for GABA release from D1 receptor-containing neurons.
Collapse
Affiliation(s)
- Carrie L Heusner
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
| | | | | | | |
Collapse
|
25
|
Gantois I, Fang K, Jiang L, Babovic D, Lawrence AJ, Ferreri V, Teper Y, Jupp B, Ziebell J, Morganti-Kossmann CM, O'Brien TJ, Nally R, Schütz G, Waddington J, Egan GF, Drago J. Ablation of D1 dopamine receptor-expressing cells generates mice with seizures, dystonia, hyperactivity, and impaired oral behavior. Proc Natl Acad Sci U S A 2007; 104:4182-7. [PMID: 17360497 PMCID: PMC1820729 DOI: 10.1073/pnas.0611625104] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Huntington's disease is characterized by death of striatal projection neurons. We used a Cre/Lox transgenic approach to generate an animal model in which D1 dopamine receptor (Drd1a)+ cells are progressively ablated in the postnatal brain. Striatal Drd1a, substance P, and dynorphin expression is progressively lost, whereas D2 dopamine receptor (Drd2) and enkephalin expression is up-regulated. Magnetic resonance spectroscopic analysis demonstrated early elevation of the striatal choline/creatine ratio, a finding associated with extensive reactive striatal astrogliosis. Sequential MRI demonstrated a progressive reduction in striatal volume and secondary ventricular enlargement confirmed to be due to loss of striatal cells. Mutant mice had normal gait and rotarod performance but displayed hindlimb dystonia, locomotor hyperactivity, and handling-induced electrographically verified spontaneous seizures. Ethological assessment identified an increase in rearing and impairments in the oral behaviors of sifting and chewing. In line with the limbic seizure profile, cell loss, astrogliosis, microgliosis, and down-regulated dynorphin expression were seen in the hippocampal dentate gyrus. This study specifically implicates Drd1a+ cell loss with tail suspension hindlimb dystonia, hyperactivity, and abnormal oral function. The latter may relate to the speech and swallowing disturbances and the classic sign of tongue-protrusion motor impersistence observed in Huntington's disease. In addition, the findings of this study support the notion that Drd1a and Drd2 are segregated on striatal projection neurons.
Collapse
Affiliation(s)
- Ilse Gantois
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
| | - Ke Fang
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
| | - Luning Jiang
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
| | - Daniela Babovic
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Andrew J. Lawrence
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
| | - Vincenzo Ferreri
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
| | - Yaroslav Teper
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
| | - Bianca Jupp
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne 3010, Australia
| | - Jenna Ziebell
- National Trauma Research Institute, Alfred Hospital, Monash University, Melbourne 3004, Australia; and
| | | | - Terence J. O'Brien
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne 3010, Australia
| | - Rachel Nally
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
| | - Günter Schütz
- Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | - John Waddington
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Gary F. Egan
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
| | - John Drago
- *Howard Florey Institute, University of Melbourne, Melbourne 3010, Australia
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
26
|
Cummings BJ, Uchida N, Tamaki SJ, Anderson AJ. Human neural stem cell differentiation following transplantation into spinal cord injured mice: association with recovery of locomotor function. Neurol Res 2006; 28:474-81. [PMID: 16808875 DOI: 10.1179/016164106x115116] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Stem cells are under intense investigation as potential therapeutics for central nervous system (CNS) injury and disease. However, several reports have suggested that stem cells grown as neurospheres and transplanted into an injured environment preferentially differentiate into astrocytes, contributing to glial scar. Further, the relationship between functional recovery and cell transplantation has not been empirically investigated in early studies. Using severe combined immunodeficient (scid) mice to minimize xenograft rejection, we report that prospectively isolated human fetal CNS-derived stem cells grown as neurospheres (hCNS-SCns) survive, migrate and express differentiation markers for neurons and oligodendrocytes after long-term engraftment in spinal cord injured (SCI) NOD-scid mice. Only rarely do these cells differentiate into glial fibrillary acidic protein (GFAP)-positive astrocytes, with no apparent contribution to glial scar. hCNS-SCns engraftment was associated with recovery of locomotor function. After long-term engraftment and stable behavioral plateaus in recovery were achieved (4 months post-transplantation), locomotor improvements were abolished by selective ablation of human cells with diphtheria toxin (DT). These data suggest that hCNS-SCns survival is required for locomotor recovery, possibly via differentiation and integration of human cells in the mouse host or continuous supply of trophic or other support necessary for gains in host cell function.
Collapse
Affiliation(s)
- Brian J Cummings
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
27
|
Abstract
Voluntary movement in animals is modulated by a number of subcortical systems. One of these resides in the basal nuclei and their associated projections and utilizes dopamine as a neurotransmitter. Apart from regulating movement, the dopaminergic axis is also involved in the control of goal-oriented behavior, cognition, and mood. Disorders of this system result in common human neurologic disorders such as Parkinson's and Huntington's diseases, as well contributing to a host of behavioral conditions, such as schizophrenia, attention deficit hyperactivity disorder, and addiction. Many individual mouse models of human dopaminergic dysfunction have been described in varying degrees of detail. However, when evaluating this region of the brain, the veterinary pathologist is confronted by a paucity of information summarizing the comparative aspects of the anatomy, physiology, and pathology of the central dopaminergic system. In this review, a systematic approach to anatomic phenotyping of the central dopaminergic system in the mouse is described and illustrated using tyrosine hydroxylase immunohistochemistry. Differences between murine neuroanatomy and comparable regions of the nonhuman primate brain are highlighted. Although the mouse is the focus of this review, conditions in domestic animals characterized by lesions within the basal nuclei and its projections are also briefly described. Murine behavioral and motor tests that accompany abnormalities of specific anatomic regions of the dopaminergic axis are summarized. Finally, we review mouse models of Parkinson's and Huntington's diseases, as well as those genetically altered mice that elucidate aspects of dopamine metabolism and receptor function.
Collapse
Affiliation(s)
- C J Zeiss
- Comparative Medicine, Yale University School of Medicine, 375 Congress Avenue, New Haven, CT 06437, USA.
| |
Collapse
|
28
|
Moussa CEH, Tomita Y, Sidhu A. Dopamine D1 receptor-mediated toxicity in human SK-N-MC neuroblastoma cells. Neurochem Int 2005; 48:226-34. [PMID: 16290264 DOI: 10.1016/j.neuint.2005.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Revised: 09/16/2005] [Accepted: 09/30/2005] [Indexed: 11/20/2022]
Abstract
Striatal degeneration occurs through unknown mechanisms in certain neurodegenerative disorders characterized by increased and sustained synaptic levels of dopamine. In the present studies, we examined the effects of treatment of SK-N-MC neuroblastoma cells with dopamine to understand the participation of dopamine D(1) receptor in postsynaptic cytotoxicity. Treatment of SK-N-MC cells either with dopamine or the D(1) receptor agonist SKF R-38393 resulted in a significant increase in the production of reactive oxygen species (by approximately 2.75-fold) and cell death ( approximately 50%), while antagonism of the D(1) receptor with SCH 23390 significantly reversed (to approximately 75% of control level) these effects. Accumulation of cAMP in dopamine treated cells (t(1/2)=1.5h) preceded changes in ionic gradient (t(1/2)=6.5h), as measured by intracellular potassium concentration and leakage of cytochrome c into the cytosol (t(1/2)=13 h), suggesting a possible staging of toxic events as a result of activation of D(1) receptor by dopamine. Examination of cellular metabolic properties with (13)C NMR spectroscopy showed an inhibitory effect on tricarboxylic acid cycle metabolism via D(1)-mediated receptors after treatment with dopamine, suggesting a direct role for D(1) receptor in dopamine-induced postsynaptic cell death. The present studies provide novel insight into a possible patho-physiological staging of cytotoxic events that are mediated by activation of D(1) receptor.
Collapse
Affiliation(s)
- Charbel E-H Moussa
- Department of Pediatrics, Georgetown University, Washington, DC 20007, USA
| | | | | |
Collapse
|
29
|
Cummings BJ, Uchida N, Tamaki SJ, Salazar DL, Hooshmand M, Summers R, Gage FH, Anderson AJ. Human neural stem cells differentiate and promote locomotor recovery in spinal cord-injured mice. Proc Natl Acad Sci U S A 2005; 102:14069-74. [PMID: 16172374 PMCID: PMC1216836 DOI: 10.1073/pnas.0507063102] [Citation(s) in RCA: 550] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We report that prospectively isolated, human CNS stem cells grown as neurospheres (hCNS-SCns) survive, migrate, and express differentiation markers for neurons and oligodendrocytes after long-term engraftment in spinal cord-injured NOD-scid mice. hCNS-SCns engraftment was associated with locomotor recovery, an observation that was abolished by selective ablation of engrafted cells by diphtheria toxin. Remyelination by hCNS-SCns was found in both the spinal cord injury NOD-scid model and myelin-deficient shiverer mice. Moreover, electron microscopic evidence consistent with synapse formation between hCNS-SCns and mouse host neurons was observed. Glial fibrillary acidic protein-positive astrocytic differentiation was rare, and hCNS-SCns did not appear to contribute to the scar. These data suggest that hCNS-SCns may possess therapeutic potential for CNS injury and disease.
Collapse
Affiliation(s)
- Brian J Cummings
- Department of Physical Medicine and Rehabilitation, Reeve-Irvine Research Center, University of California, Irvine, CA 92697, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Chen J, Rusnak M, Luedtke RR, Sidhu A. D1 Dopamine Receptor Mediates Dopamine-induced Cytotoxicity via the ERK Signal Cascade. J Biol Chem 2004; 279:39317-30. [PMID: 15247297 DOI: 10.1074/jbc.m403891200] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Postsynaptic striatal neurodegeneration occurs through unknown mechanisms, but it is linked to high extracellular levels of synaptic dopamine. Dopamine-mediated cytotoxicity of striatal neurons occurs through two distinct pathways: autoxidation and the D1 dopamine receptor-linked signaling pathway. Here we investigated the mitogen-activated protein kinase (MAPK) signaling pathways activated upon the acute stimulation of D1 dopamine receptors. In SK-N-MC neuroblastoma cells, endogenously expressing D1 dopamine receptors, dopamine caused activation of phosphorylated (p-)ERK1/2 and of the stress-signaling kinases, p-JNK and p-p38 MAPK, in a time- and dose-dependent manner. Selective stimulation of D1 receptors with the agonist SKF R-38393 caused p-ERK1/2, but not p-JNK or p-p38 MAPK activation, in a manner sensitive to the receptor-selective antagonist SCH 23390, protein kinase A inhibition (KT5720), and MEK1/2 inhibition (U0126 or PD98059). Activation of ERK by D1 dopamine receptors resulted in oxidative stress and cytotoxicity. In cells transfected with a catalytically defective mutant of MEK1, the upstream ERK-specific kinase, both dopamine- and SKF R-38393-mediated cytotoxicity was markedly attenuated, confirming the participation of the ERK signaling pathway. Cell fractionation studies showed that only a small amount of p-ERK1/2 was translocated to the nucleus, with the majority retained in the cytoplasm. From coimmunoprecipitation studies, p-ERK was found to form stable heterotrimeric complexes with the D1 dopamine receptor and beta-arrestin2. In cells transfected with the dominant negative mutant of beta-arrestin2, the formation of such complexes was substantially inhibited. These data provide novel mechanistic insights into the role of ERK in the cytotoxicity mediated upon activation of the D1 dopamine receptor.
Collapse
Affiliation(s)
- Jun Chen
- Department of Pediatrics, Georgetown University, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
31
|
Wersinger C, Chen J, Sidhu A. Bimodal induction of dopamine-mediated striatal neurotoxicity is mediated through both activation of D1 dopamine receptors and autoxidation. Mol Cell Neurosci 2004; 25:124-37. [PMID: 14962746 DOI: 10.1016/j.mcn.2003.10.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2003] [Revised: 10/02/2003] [Accepted: 10/02/2003] [Indexed: 10/26/2022] Open
Abstract
Striatal neurodegeneration occurs through unknown mechanisms in certain neurodegenerative disorders characterized by increased and sustained synaptic levels of dopamine (DA). Treatment of rat primary striatal neurons with DA causes profound neurotoxicity, with increased production of free radicals and accelerated neuronal death. DA effects were partly reduced by the antioxidant sodium metabisulfite (SMBS), and the D1 DA receptor antagonist, SCH 23390, and were completely blocked upon co-treatment with SMBS and SCH 23390. Part of DA effects were mimicked by either H(2)O(2), or by the D1 agonist, SKF R-38393, indicating the existence of two distinct signaling pathways through which the neurotoxicity of DA is manifest. DA effects did not proceed through D2-like DA or beta-adrenergic receptor signaling pathways. The D1 receptor-mediated and the autoxidative pathways of DA neurotoxicity converge to cause activation and/or increased synthesis of neuronal and inducible, but not endothelial, nitric oxide synthase (NOS). The reduction of DA striatal neurotoxicity through blockade of D1 DA receptors, suggests novel therapeutic approaches in the management of striatal neurodegeneration.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antioxidants/pharmacology
- Basal Ganglia Diseases/metabolism
- Basal Ganglia Diseases/physiopathology
- Cells, Cultured
- Dopamine/metabolism
- Dopamine/toxicity
- Dopamine Agonists/pharmacology
- Dopamine Antagonists/pharmacology
- Hydrogen Peroxide/pharmacology
- Neostriatum/metabolism
- Neostriatum/physiopathology
- Nerve Degeneration/chemically induced
- Nerve Degeneration/metabolism
- Nerve Degeneration/physiopathology
- Neurons/drug effects
- Neurons/metabolism
- Neurotoxins/metabolism
- Neurotoxins/toxicity
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type I
- Nitric Oxide Synthase Type II
- Oxidants/pharmacology
- Oxidative Stress/drug effects
- Oxidative Stress/physiology
- Rats
- Receptors, Adrenergic, beta/drug effects
- Receptors, Adrenergic, beta/metabolism
- Receptors, Dopamine D1/drug effects
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/drug effects
- Receptors, Dopamine D2/metabolism
Collapse
|
32
|
Chen J, Wersinger C, Sidhu A. Chronic stimulation of D1 dopamine receptors in human SK-N-MC neuroblastoma cells induces nitric-oxide synthase activation and cytotoxicity. J Biol Chem 2003; 278:28089-100. [PMID: 12738794 DOI: 10.1074/jbc.m303094200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Elevated synaptic levels of dopamine may induce striatal neurodegeneration in l-DOPA-unresponsive parkinsonism subtype of multiple system atrophy (MSA-P subtype), multiple system atrophy, and methamphetamine addiction. We examined the participation of dopamine and D1 dopamine receptors in the genesis of postsynaptic neurodegeneration. Chronic treatment of human SK-N-MC neuroblastoma cells with dopamine or H2O2 increased NO production and accelerated cytotoxicity, as indexed by enhanced nitrite levels and cell death. The antioxidant sodium metabisulfite or SCH 23390, a D1 dopamine receptor-selective antagonist, partially blocked dopamine effects but together ablated dopamine-mediated cytotoxicity, indicating the participation of both autoxidation and D1 receptor stimulation. Direct activation of D1 dopamine receptors with SKF R-38393 caused cytotoxicity, which was refractory to sodium metabisulfite. Dopamine and SKF R-38393 induced overexpression of the nitric-oxide synthase (NOS) isoforms neuronal NOS, inducible NOS (iNOS), and endothelial NOS in a protein kinase A-dependent manner. Functional studies showed that approximately 60% of total NOS activity was due to activation of iNOS. The NOS inhibitor N(G)-nitro-l-arginine methyl ester and genistein, wortmannin, or NF-kappaB SN50, inhibitors of protein tyrosine kinases phosphatidylinositol 3-kinase and NF-kappaB, respectively, reduced nitrite production by dopamine and SKF R-38393 but were less effective in attenuating H2O2-mediated effects. In rat striatal neurons, dopamine and SKF R-38393, but not H2O2, accelerated cell death through increased expression of neuronal NOS and iNOS but not endothelial NOS. These data demonstrate a novel pathway of dopamine-mediated postsynaptic oxidative stress and cell death through direct activation of NOS enzymes by D1 dopamine receptors and its associated signaling pathways.
Collapse
Affiliation(s)
- Jun Chen
- Department of Pediatrics, Georgetown University, Washington, DC 20007, USA
| | | | | |
Collapse
|
33
|
Wong JYF, Clifford JJ, Massalas JS, Finkelstein DI, Horne MK, Waddington JL, Drago J. Neurochemical changes in dopamine D1, D3 and D1/D3 receptor knockout mice. Eur J Pharmacol 2003; 472:39-47. [PMID: 12860471 DOI: 10.1016/s0014-2999(03)01862-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Neurochemical changes were examined in dopamine D1 receptor knockout (D1(-/-)), dopamine D3 receptor knockout (D3(-/-)) and dopamine D1/D3 receptor double knockout (D1(-/-)D3(-/-)) mice. The level of dopamine D1- and D2-like receptors and gamma-aminobutyric acid (GABA(A)) receptor was assessed by ligand autoradiography and dopamine D1- and D2 receptor, enkephalin, dynorphin and substance P transcripts measured by in situ hybridization. D1(-/-) mice had normal GABA(A) receptor levels, reduced dynorphin and substance P, and increased enkephalin mRNA and dopamine D2-like binding. D1(-/-)D3(-/-) mice evidenced decreased dynorphin and substance P but normal enkephalin expression, whereas dopamine D2-like and GABA(A) receptor binding were increased. Major changes occur in substance P and dynorphin expression in D1(-/-) mice and these changes are unaffected by loss of dopamine D3 receptors. Upregulated dopamine D2-like binding and enkephalin in D1(-/-) mice may be due to decreased dopamine turnover. Upregulated enkephalin in D1(-/-) mice is dependent on functional dopamine D3 receptors.
Collapse
MESH Headings
- Animals
- Autoradiography
- Dopamine Plasma Membrane Transport Proteins
- Dynorphins/biosynthesis
- Dynorphins/metabolism
- Enkephalins/biosynthesis
- Enkephalins/metabolism
- Ligands
- Male
- Membrane Glycoproteins
- Membrane Transport Proteins/metabolism
- Mice
- Mice, Knockout
- Nerve Tissue Proteins
- Protein Binding
- RNA, Messenger/biosynthesis
- Receptors, Dopamine D1/biosynthesis
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/biosynthesis
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Receptors, Dopamine D3
- Receptors, GABA-A/metabolism
- Receptors, Muscarinic/metabolism
- Substance P/metabolism
- Telencephalon/metabolism
Collapse
Affiliation(s)
- John Y F Wong
- Neurosciences Group, Monash University, Department of Medicine, Monash Medical Centre, Level 5 Block E, 246 Clayton Road, Clayton, Victoria 3168, Australia
| | | | | | | | | | | | | |
Collapse
|
34
|
Tan S, Hermann B, Borrelli E. Dopaminergic mouse mutants: investigating the roles of the different dopamine receptor subtypes and the dopamine transporter. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 54:145-97. [PMID: 12785287 DOI: 10.1016/s0074-7742(03)54005-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Shirlee Tan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, BP 10142 Illkirch, C.U. de Strasbourg, France
| | | | | |
Collapse
|
35
|
Kiehn O, Butt SJB. Physiological, anatomical and genetic identification of CPG neurons in the developing mammalian spinal cord. Prog Neurobiol 2003; 70:347-61. [PMID: 12963092 DOI: 10.1016/s0301-0082(03)00091-1] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The basic motor patterns underlying rhythmic limb movements during locomotion are generated by neuronal networks located within the spinal cord. These networks are called Central Pattern Generators (CPGs). Isolated spinal cord preparations from newborn rats and mice have become increasingly important for understanding the organization of the CPG in the mammalian spinal cord. Early studies using these preparations have focused on the overall network structure and the localization of the CPG. In this review we concentrate on recent experiments aimed at identifying and characterizing CPG-interneurons in the rodent. These experiments include the organization and function of descending commissural interneurons (dCINs) in the hindlimb CPG of the neonatal rat, as well as the role of Ephrin receptor A4 (EphA4) and its Ephrin ligand B3 (EphrinB3), in the construction of the mammalian locomotor network. These latter experiments have defined EphA4 as a molecular marker for mammalian excitatory hindlimb CPG neurons. We also review genetic approaches that can be applied to the mouse spinal cord. These include methods for identifying sub-populations of neurons by genetically encoded reporters, techniques to trace network connectivity with cell-specific genetically encoded tracers, and ways to selectively ablate or eliminate neuron populations from the CPG. We propose that by applying a multidisciplinary approach it will be possible to understand the network structure of the mammalian locomotor CPG. Such an understanding will be instrumental in devising new therapeutic strategies for patients with spinal cord injury.
Collapse
Affiliation(s)
- Ole Kiehn
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 171 77 Stockholm, Sweden.
| | | |
Collapse
|
36
|
Abstract
The targeted modification of the mammalian genome has a variety of applications in research, medicine, and biotechnology. Site-specific recombinases have become significant tools in all of these areas. Conditional gene targeting using site-specific recombinases has enabled the functional analysis of genes, which cannot be inactivated in the germline. The site-specific integration of adeno-associated virus, a major gene therapy vehicle, relies on the recombinase activity of the viral rep proteins. Site-specific recombinases also allow the precise integration of open reading frames encoding pharmaceutically relevant proteins into highly active gene loci in cell lines and transgenic animals. These goals have been accomplished by using a variety of genetic strategies but only a few recombinase proteins. However, the vast repertoire of recombinases, which has recently become available as a result of large-scale sequencing projects, may provide a rich source for the development of novel strategies to precisely alter mammalian genomes.
Collapse
Affiliation(s)
- Andreas F Kolb
- Cell Physiology Group, Hannah Research Institute, Ayr, United Kingdom.
| |
Collapse
|
37
|
Nadeau H, Lester HA. NRSF causes cAMP-sensitive suppression of sodium current in cultured hippocampal neurons. J Neurophysiol 2002; 88:409-21. [PMID: 12091564 DOI: 10.1152/jn.2002.88.1.409] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The neuron restrictive silencer factor (NRSF/REST) has been shown to bind to the promoters of many neuron-specific genes and is able to suppress transcription of Na(+) channels in PC12 cells, although its functional effect in terminally differentiated neurons is unknown. We constructed lentiviral vectors to express NRSF as a bicistronic message with green fluorescent protein (GFP) and followed infected hippocampal neurons in culture over a period of 1-2 wk. NRSF-expressing neurons showed a time-dependent suppression of Na(+) channel function as measured by whole cell electrophysiology. Suppression was reversed or prevented by the addition of membrane-permeable cAMP analogues and enhanced by cAMP antagonists but not affected by increasing protein expression with a viral enhancer. Secondary effects, including altered sensitivity to glutamate and GABA and reduced outward K(+) currents, were duplicated by culturing GFP-infected control neurons in TTX. The striking similarity of the phenotypes makes NRSF potentially useful as a genetic "silencer" and also suggests avenues of further exploration that may elucidate the transcription factor's in vivo role in neuronal plasticity.
Collapse
Affiliation(s)
- H Nadeau
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA.
| | | |
Collapse
|
38
|
Clifford JJ, Drago J, Natoli AL, Wong JYF, Kinsella A, Waddington JL, Vaddadi KS. Essential fatty acids given from conception prevent topographies of motor deficit in a transgenic model of Huntington's disease. Neuroscience 2002; 109:81-8. [PMID: 11784701 DOI: 10.1016/s0306-4522(01)00409-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Transgenic R6/1 mice incorporate a human genomic fragment containing promoter elements exon 1 and a portion of intron 2 of the Huntingtin gene responsible for Huntington's disease. They develop late-onset neurological deficits in a manner similar to the motor abnormalities of the disorder. As essential fatty acids are phospholipid components of cell membranes which may influence cell death and movement disorder phenotype, R6/1 and normal mice were randomised to receive a mixture of essential fatty acids or placebo on alternate days throughout life. Over mid-adulthood, topographical assessment of behaviour revealed R6/1 transgenics to evidence progressive shortening of stride length, with progressive reductions in locomotion, elements of rearing, sniffing, sifting and chewing, and an increase in grooming. These deficits were either not evident or materially diminished in R6/1 transgenics receiving essential fatty acids. R6/1 transgenics also showed reductions in body weight and in brain dopamine D(1)-like and D(2)-like quantitative receptor autoradiography which were unaltered by essential fatty acids.These findings indicate that early and sustained treatment with essential fatty acids are able to protect against motor deficits in R6/1 transgenic mice expressing exon 1 and a portion of intron 2 of the Huntingtin gene, and suggest that essential fatty acids may have therapeutic potential in Huntington's disease.
Collapse
Affiliation(s)
- J J Clifford
- Department of Clinical Pharmacology, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland.
| | | | | | | | | | | | | |
Collapse
|
39
|
Lee EJ, Jameson JL. Cell-specific Cre-mediated activation of the diphtheria toxin gene in pituitary tumor cells: potential for cytotoxic gene therapy. Hum Gene Ther 2002; 13:533-42. [PMID: 11874631 DOI: 10.1089/10430340252809829] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diphtheria toxin has been suggested for the treatment of malignant cancer. In this paper, we describe a strategy for targeting the expression of the diphtheria toxin gene to growth hormone (GH)-producing pituitary tumor cells using adenoviral vectors. We generated adenoviral vectors in which a stuffer DNA fragment, flanked by two loxP sequences, was placed between the GH or cytomegalovirus (CMV) promoters and the diphtheria toxin gene (GH-loxP-DT, and CMV-loxP-DT) or the beta-Gal gene (GH-loxP-Gal, and CMV-loxP-Gal). Co-infection of GH-loxP-DT with either CMV-Cre or GH-Cre induced cytotoxicity that was limited to GH4 cells. Little or no cytopathic effect was seen in GH4 cells infected with control viruses (CMV-loxP-Gal or GH-loxP-Gal with CMV-Cre or GH-Cre). To test the effectiveness of this strategy in vivo, GH4 cells were transplanted into nude mice. Intratumoral co-injection of adenoviruses carrying diphtheria toxin (GH-loxP-DT, and CMV-loxP-DT) and Cre recombinase (GH-Cre, and CMV-Cre) caused rapid regression of the transplanted GH4 tumors. These results indicate that Cre-mediated activation of a loxP-repressed form of the DT gene provides a useful strategy for targeted suicide gene therapy. This approach may be useful for GH-secreting adenomas and should be applicable to other neoplastic disorders.
Collapse
Affiliation(s)
- Eun Jig Lee
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Medical School, Chicago, IL 60611-2908, USA
| | | |
Collapse
|
40
|
Abstract
One of the most powerful tools that the molecular biology revolution has given us is the ability to turn genes on and off at our discretion. In the mouse, this has been accomplished by using binary systems in which gene expression is dependent on the interaction of two components, resulting in either transcriptional transactivation or DNA recombination. During recent years, these systems have been used to analyse complex and multi-staged biological processes, such as embryogenesis and cancer, with unprecedented precision. Here, I review these systems and discuss certain studies that exemplify the advantages and limitations of each system.
Collapse
Affiliation(s)
- M Lewandoski
- Section of Genetics of Vertebrate Development, Laboratory of Cancer and Developmental Biology, National Cancer Institute-Frederick Cancer Research and Development Center, Frederick, Maryland 21702-1201, USA.
| |
Collapse
|
41
|
Wells T, Carter DA. Genetic engineering of neural function in transgenic rodents: towards a comprehensive strategy? J Neurosci Methods 2001; 108:111-30. [PMID: 11478971 DOI: 10.1016/s0165-0270(01)00391-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
As mammalian genome projects move towards completion, the attention of molecular neuroscientists is currently moving away from gene identification towards both cell-specific gene expression patterns (neuronal transcriptions) and protein expression/interactions (neuronal proteomics). In the long term, attention will increasingly be directed towards experimental interventions which are able to question neuronal function in a sophisticated manner that is cognisant of both transcriptomic and proteomic organization. Central to this effort will be the application of a new generation of transgenic approaches which are now evolving towards an appropriate level of molecular, temporal and spatial resolution. In this review, we summarize recent developments in transgenesis, and show how they have been applied in the principal model species for neuroscience, namely rats and mice. Current concepts of transgene design are also considered together with an overview of new genetically-encoded tools including both cellular indicators such as fluorescent activity reporters, and cellular regulators such as dominant negative signalling factors. Application of these tools in a whole animal context can be used to question both basic concepts of brain function, and also current concepts of underlying dysfuction in neurological diseases.
Collapse
Affiliation(s)
- T Wells
- School of Biosciences, Cardiff University, PO Box 911, Museum Avenue, Cardiff CF10 3US, UK
| | | |
Collapse
|
42
|
Snyder-Keller A, Costantini LC, Graber DJ. Development of striatal patch/matrix organization in organotypic co-cultures of perinatal striatum, cortex and substantia nigra. Neuroscience 2001; 103:97-109. [PMID: 11311790 DOI: 10.1016/s0306-4522(00)00535-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Organotypic cultures of fetal or early postnatal striatum were used to assess striatal patch formation and maintenance in the presence or absence of dopaminergic and glutamatergic influences. Vibratome-cut slices of the striatum prepared from embryonic day 19 to postnatal day 4 rat pups were maintained in static culture on clear membrane inserts in Dulbecco's modified Eagle's medium/F12 (1:1) with 20% horse serum. Some were co-cultured with embryonic day 12-16 ventral mesencephalon and/or embryonic day 19 to postnatal day 4 cortex, which produced a dense dopaminergic innervation and a modest cortical innervation. Donors of striatal and cortical tissue were previously injected with bromo-deoxyuridine (BrdU) on embryonic days 13 and 14 in order to label striatal neurons destined to populate the patch compartment of the striatum. Patches of BrdU-immunoreactive cells were maintained in organotypic cultures of late prenatal (embryonic days 20-22) or early postnatal striatum in the absence of nigral dopaminergic or cortical glutamatergic influences. In slices taken from embryonic day 19 fetuses prior to the time of in vivo patch formation, patches were observed to form after 10 days in vitro, in 39% of nigral-striatal co-cultures compared to 6% of striatal slices cultured alone or in the presence of cortex only. Patches of dopaminergic fibers, revealed by tyrosine hydroxylase immunoreactivity, were observed in the majority of nigral-striatal co-cultures. Immunostaining for the AMPA-type glutamate receptor GluR1 revealed a dense patch distribution in nearly all cultures, which developed in embryonic day 19 cultures after at least six days in vitro. These findings indicate that striatal patch/matrix organization is maintained in organotypic culture, and can be induced to form in vitro in striatal slices removed from fetuses prior to the time of in vivo patch formation. Furthermore, dopaminergic innervation from co-cultured pieces of ventral mesencephalon enhances patch formation in organotypic cultures.
Collapse
Affiliation(s)
- A Snyder-Keller
- Wadsworth Center, New York State Department of Health, P.O. Box 509, Empire State Plaza, Albany, NY 12201, USA.
| | | | | |
Collapse
|
43
|
Waddington JL, Clifford JJ, McNamara FN, Tomiyama K, Koshikawa N, Croke DT. The psychopharmacology-molecular biology interface: exploring the behavioural roles of dopamine receptor subtypes using targeted gene deletion ('knockout'). Prog Neuropsychopharmacol Biol Psychiatry 2001; 25:925-64. [PMID: 11383985 DOI: 10.1016/s0278-5846(01)00152-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the absence of selective agonists and antagonists able to discriminate between individual members of the D1-like and D2-like families of dopamine receptor subtypes, functional parcellation has remained problematic. 'Knockout' of these subtypes by targeted gene deletion offers a new approach to evaluating their roles in the regulation of behaviour. Like any new technique, 'knockout' has associated with it a number of methodological limitations that are now being addressed in a systematic manner. Studies on the phenotype of D1(A/1), D(1B/5), D2, D3 and D4 'knockouts' at the level of spontaneous and agonist/antagonist-induced behaviour are reviewed, in terms of methodological issues, neuronal implications and potential clinical relevance. Dopamine receptor subtype 'knockout' is a nascent technology that is now beginning to fulfil its potential. It is being complemented by more systematic phenotypic characterisation at the level of behaviour and additional, molecular biologically-based approaches.
Collapse
Affiliation(s)
- J L Waddington
- Department of Clinical Pharmacology, Royal College of Surgeons in Ireland, Dublin
| | | | | | | | | | | |
Collapse
|
44
|
Padungchaichot P, Wong JY, Natoli AL, Massalas JS, Finkelstein DI, Lawrence AL, Drago J. Early direct and transneuronal effects in mice with targeted expression of a toxin gene to D1 dopamine receptor neurons. Neuroscience 2000; 95:1025-33. [PMID: 10682709 DOI: 10.1016/s0306-4522(99)00497-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The neurochemical profile was examined at postnatal day 3-4 in mutant mice generated by in vivo Cre mediated activation of an attenuated diphtheria toxin gene inserted into the D1 dopamine receptor gene locus. An earlier study of this model had shown that D1 dopamine receptor, substance P and dynorphin were not expressed in the striatum. Quantitative in situ hybridization analysis showed an increase in D2 dopamine receptor and enkephalin messenger RNA expression. The nigrostriatal pathway in the mutant pups was intact with a normal number of dopaminergic neurons in the substantia nigra and the ventral tegmental area in addition to a normal pattern of striatal dopamine transporter and tyrosine hydroxylase immunoreactivity. Quantitative analysis of striatal dopamine transporter density using [3H]mazindol showed a reduction of 26% suggesting a degree of transneuronal down-regulation. There was also a 49% reduction of striatal GABA receptor binding and a 36% reduction of striatal muscarinic receptor binding in mutant pups. The number of healthy striatal neuropeptide Y-containing interneurons was also substantially down-regulated in the mutant striatum. In contrast, there was an increase in the number of striatal cholinergic interneurons. Down-regulated cortical GABA receptor and muscarinic receptor binding was also observed in addition to subtle morphological changes in the neuropeptide Y-expressing population of cortical neurons. The changes reflect the early cascade of events which follows the ablation of D1 dopamine receptor-positive cells. Although extensive changes in a number of striatal and cortical neurons were demonstrated, only subtle transneuronal effects were seen in the nigrostriatal pathway.
Collapse
Affiliation(s)
- P Padungchaichot
- Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
45
|
Wong JY, Padungchaichot P, Massalas JS, Drago J. Late direct and transneuronal effects in mice with targeted expression of a toxin gene to D1 dopamine receptor neurons. Neuroscience 2000; 95:1035-41. [PMID: 10682710 DOI: 10.1016/s0306-4522(99)00498-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Detailed analysis of a novel transgenic model of basal ganglia disease has been undertaken. In this model the expression of an attenuated form of the diphtheria toxin gene was tightly controlled by D1 dopamine receptor regulatory domains. The behavioral and both direct toxin-mediated and transneuronal effects observed in pups in the first postnatal week have been described. Although younger pups are bradykinetic, older pups have a hyperkinetic syndrome with gait abnormality, postural instability and myoclonic jerks typical of human basal ganglia diseases such as Huntington's disease. As expected, striatal D1 dopamine receptor, dynorphin and substance P transcripts were not detected by in situ hybridization but there was a 27% increase in striatal D2 dopamine receptor messenger RNA and a 65% increase in enkephalin messenger RNA expression. Receptor autoradiographic studies confirmed the lack of D1-class binding in the mutant striatum and in contrast to young pups, a substantial increase in striatal D2-class binding. Autoradiographic quantitation also showed a 30% increase in striatal dopamine transporter binding. In addition to the changes described in the striatopallidal and nigrostriatal pathways, up-regulated dynorphin and substance P messenger RNA expression was also seen in the cortex. The capacity of the developing brain for neurochemical adaptation following injury is dramatic. The results show that primary loss of D1 dopamine receptor-positive striatonigral pathway neurons is sufficient to generate a hyperkinetic phenotype.
Collapse
Affiliation(s)
- J Y Wong
- Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|