Administrative Appointments


  • Department of Neurosurgery, and Department of Neurology and Neurological Sciences, by courtesy, Stanford University School of Medicine (2012 - Present)

Honors & Awards


  • K99/R00 Pathway to Independence Award, NIH/NINDS (2011)
  • Postdoctoral Fellowship, Parkinson’s Disease Foundation (2011)
  • Klingenstein Fellowship Awards in Neuroscience, Klingenstein Foundation (2013)
  • Kavli Fellow, Kavli Foundation (2014)

Professional Education


  • Ph.D., Interdepartmental Neuroscience PhD Program , Department of Physiology, Northwestern University, Neuroscience (2007)

Current Research and Scholarly Interests


The interplay between motor cortex, sensory cortex, thalamus and basal ganglia is essential for neural computations involved in generating voluntary movements. Our goal is to dissect the functional organization of motor circuits, particularly cortico-thalamo-basal ganglia networks, using electrophysiology, 2-photon microscopy, optogenetics, and genetic tools. The long-term scientific goal of the lab is to construct functional circuit diagrams and establish causal relationships between activity in specific groups of neurons, circuit function, animal motor behavior and motor learning, and thereby to decipher how the basal ganglia process information and guide motor behavior. We will achieve this by investigating the synaptic organization and function that involve the cortex, thalamus and basal ganglia at the molecular, cellular and circuit level. Currently, we are focusing on several questions:
How are excitatory and inhibitory inputs integrated in the striatum?
How do feed-forward and recurrent local inhibitions balance the excitation in the striatum?
How are functional maps modulated in motor behavior and motor learning?
Our goal is to bridge the gap between molecular or cellular events and the circuit mechanisms that underlie motor behavior. In addition, we aim to further help construct the details of psychomotor disorder ‘circuit diagrams,’ such as the pathophysiological changes in Parkinson’s disease.

2018-19 Courses


Stanford Advisees


Graduate and Fellowship Programs


All Publications


  • The THO Complex Coordinates Transcripts for Synapse Development and Dopamine Neuron Survival. Cell Maeder, C. I., Kim, J., Liang, X., Kaganovsky, K., Shen, A., Li, Q., Li, Z., Wang, S., Xu, X. Z., Li, J. B., Xiang, Y. K., Ding, J. B., Shen, K. 2018

    Abstract

    Synaptic vesicle and active zone proteins are required for synaptogenesis. The molecular mechanisms for coordinated synthesis of these proteins are not understood. Using forward genetic screens, we identified the conserved THO nuclear export complex (THOC) as an important regulator of presynapse development in C.elegans dopaminergic neurons. In THOC mutants, synaptic messenger RNAs are retained in the nucleus, resulting in dramatic decrease of synaptic protein expression, near complete loss of synapses, and compromised dopamine function. CRE binding protein (CREB) interacts with THOC to mark synaptic transcripts for efficient nuclear export. Deletion of Thoc5, a THOC subunit, in mouse dopaminergic neurons causes severe defects in synapse maintenance and subsequent neuronal death in the substantia nigra compacta. These cellular defects lead to abrogated dopamine release, ataxia, and animal death. Together, our results argue that nuclear export mechanisms can select specific mRNAs and be a rate-limiting step for neuronal differentiation and survival.

    View details for DOI 10.1016/j.cell.2018.07.046

    View details for PubMedID 30146163

  • Cell-type-specific inhibition of the dendritic plateau potential in striatal spiny projection neurons. Proceedings of the National Academy of Sciences of the United States of America Du, K., Wu, Y. W., Lindroos, R., Liu, Y., Rózsa, B., Katona, G., Ding, J. B., Kotaleski, J. H. 2017; 114 (36): E7612–E7621

    Abstract

    Striatal spiny projection neurons (SPNs) receive convergent excitatory synaptic inputs from the cortex and thalamus. Activation of spatially clustered and temporally synchronized excitatory inputs at the distal dendrites could trigger plateau potentials in SPNs. Such supralinear synaptic integration is crucial for dendritic computation. However, how plateau potentials interact with subsequent excitatory and inhibitory synaptic inputs remains unknown. By combining computational simulation, two-photon imaging, optogenetics, and dual-color uncaging of glutamate and GABA, we demonstrate that plateau potentials can broaden the spatiotemporal window for integrating excitatory inputs and promote spiking. The temporal window of spiking can be delicately controlled by GABAergic inhibition in a cell-type-specific manner. This subtle inhibitory control of plateau potential depends on the location and kinetics of the GABAergic inputs and is achieved by the balance between relief and reestablishment of NMDA receptor Mg2+ block. These findings represent a mechanism for controlling spatiotemporal synaptic integration in SPNs.

    View details for DOI 10.1073/pnas.1704893114

    View details for PubMedID 28827326

    View details for PubMedCentralID PMC5594658

  • Aldehyde dehydrogenase 1a1 mediates a GABA synthesis pathway in midbrain dopaminergic neurons. Science Kim, J., Ganesan, S., Luo, S. X., Wu, Y., Park, E., Huang, E. J., Chen, L., Ding, J. B. 2015; 350 (6256): 102-106

    Abstract

    Midbrain dopamine neurons are an essential component of the basal ganglia circuitry, playing key roles in the control of fine movement and reward. Recently, it has been demonstrated that γ-aminobutyric acid (GABA), the chief inhibitory neurotransmitter, is co-released by dopamine neurons. Here, we show that GABA co-release in dopamine neurons does not use the conventional GABA-synthesizing enzymes, glutamate decarboxylases GAD65 and GAD67. Our experiments reveal an evolutionarily conserved GABA synthesis pathway mediated by aldehyde dehydrogenase 1a1 (ALDH1a1). Moreover, GABA co-release is modulated by ethanol (EtOH) at concentrations seen in blood alcohol after binge drinking, and diminished ALDH1a1 leads to enhanced alcohol consumption and preference. These findings provide insights into the functional role of GABA co-release in midbrain dopamine neurons, which may be essential for reward-based behavior and addiction.

    View details for DOI 10.1126/science.aac4690

    View details for PubMedID 26430123

  • Dynamic rewiring of neural circuits in the motor cortex in mouse models of Parkinson's disease. Nature neuroscience Guo, L., Xiong, H., Kim, J., Wu, Y., Lalchandani, R. R., Cui, Y., Shu, Y., Xu, T., Ding, J. B. 2015; 18 (9): 1299-1309

    Abstract

    Dynamic adaptations in synaptic plasticity are critical for learning new motor skills and maintaining memory throughout life, which rapidly decline with Parkinson's disease (PD). Plasticity in the motor cortex is important for acquisition and maintenance of motor skills, but how the loss of dopamine in PD leads to disrupted structural and functional plasticity in the motor cortex is not well understood. Here we used mouse models of PD and two-photon imaging to show that dopamine depletion resulted in structural changes in the motor cortex. We further discovered that dopamine D1 and D2 receptor signaling selectively and distinctly regulated these aberrant changes in structural and functional plasticity. Our findings suggest that both D1 and D2 receptor signaling regulate motor cortex plasticity, and loss of dopamine results in atypical synaptic adaptations that may contribute to the impairment of motor performance and motor memory observed in PD.

    View details for DOI 10.1038/nn.4082

    View details for PubMedID 26237365

  • Input- and Cell-Type-Specific Endocannabinoid-Dependent LTD in the Striatum. Cell reports Wu, Y., Kim, J., Tawfik, V. L., Lalchandani, R. R., Scherrer, G., Ding, J. B. 2015; 10 (1): 75-87

    Abstract

    Changes in basal ganglia plasticity at the corticostriatal and thalamostriatal levels are required for motor learning. Endocannabinoid-dependent long-term depression (eCB-LTD) is known to be a dominant form of synaptic plasticity expressed at these glutamatergic inputs; however, whether eCB-LTD can be induced at all inputs on all striatal neurons is still debatable. Using region-specific Cre mouse lines combined with optogenetic techniques, we directly investigated and distinguished between corticostriatal and thalamostriatal projections. We found that eCB-LTD was successfully induced at corticostriatal synapses, independent of postsynaptic striatal spiny projection neuron (SPN) subtype. Conversely, eCB-LTD was only nominally present at thalamostriatal synapses. This dichotomy was attributable to the minimal expression of cannabinoid type 1 (CB1) receptors on thalamostriatal terminals. Furthermore, coactivation of dopamine receptors on SPNs during LTD induction re-established SPN-subtype-dependent eCB-LTD. Altogether, our findings lay the groundwork for understanding corticostriatal and thalamostriatal synaptic plasticity and for striatal eCB-LTD in motor learning.

    View details for DOI 10.1016/j.celrep.2014.12.005

    View details for PubMedID 25543142

    View details for PubMedCentralID PMC4286501

  • Balanced Activity between Kv3 and Nav Channels Determines Fast-Spiking in Mammalian Central Neurons. iScience Gu, Y., Servello, D., Han, Z., Lalchandani, R. R., Ding, J. B., Huang, K., Gu, C. 2018; 9: 120–37

    Abstract

    Fast-spiking (FS) neurons can fire action potentials (APs) up to 1,000Hz and play key roles in vital functions such as sound location, motor coordination, and cognition. Here we report that the concerted actions of Kv3 voltage-gated K+ (Kv) and Na+ (Nav) channels are sufficient and necessary for inducing and maintaining FS. Voltage-clamp analysis revealed a robust correlation between the Kv3/Nav current ratio and FS. Expressing Kv3 channels alone could convert 30%-60% slow-spiking (SS) neurons to FS in culture. In contrast, co-expression of either Nav1.2 or Nav1.6 together with Kv3.1 or Kv3.3, but not alone or with Kv1.2, converted SS to FS with 100% efficiency. Furthermore, RNA-sequencing-based genome-wide analysis revealed that the Kv3/Nav ratio and Kv3 expression levels strongly correlated with the maximal AP frequencies. Therefore, FS is established by the properly balanced activities of Kv3 and Nav channels and could be further fine-tuned by channel biophysical features and localization patterns.

    View details for DOI 10.1016/j.isci.2018.10.014

    View details for PubMedID 30390433

  • Diametric neural ensemble dynamics in parkinsonian and dyskinetic states. Nature Parker, J. G., Marshall, J. D., Ahanonu, B., Wu, Y. W., Kim, T. H., Grewe, B. F., Zhang, Y., Li, J. Z., Ding, J. B., Ehlers, M. D., Schnitzer, M. J. 2018

    Abstract

    Loss of dopamine in Parkinson's disease is hypothesized to impede movement by inducing hypo- and hyperactivity in striatal spiny projection neurons (SPNs) of the direct (dSPNs) and indirect (iSPNs) pathways in the basal ganglia, respectively. The opposite imbalance might underlie hyperkinetic abnormalities, such as dyskinesia caused by treatment of Parkinson's disease with the dopamine precursor L-DOPA. Here we monitored thousands of SPNs in behaving mice, before and after dopamine depletion and during L-DOPA-induced dyskinesia. Normally, intermingled clusters of dSPNs and iSPNs coactivated before movement. Dopamine depletion unbalanced SPN activity rates and disrupted the movement-encoding iSPN clusters. Matching their clinical efficacy, L-DOPA or agonism of the D2 dopamine receptor reversed these abnormalities more effectively than agonism of the D1 dopamine receptor. The opposite pathophysiology arose in L-DOPA-induced dyskinesia, during which iSPNs showed hypoactivity and dSPNs showed unclustered hyperactivity. Therefore, both the spatiotemporal profiles and rates of SPN activity appear crucial to striatal function, and next-generation treatments for basal ganglia disorders should target both facets of striatal activity.

    View details for DOI 10.1038/s41586-018-0090-6

    View details for PubMedID 29720658

  • A cell-type-specific jolt for motor disorders. Nature neuroscience Wu, Y., Ding, J. B. 2017; 20 (6): 763-765

    View details for DOI 10.1038/nn.4565

    View details for PubMedID 28542150

  • Motor Learning in Animal Models of Parkinson's Disease: Aberrant Synaptic Plasticity in the Motor Cortex MOVEMENT DISORDERS Xu, T., Wang, S., Lalchandani, R. R., Ding, J. B. 2017; 32 (4): 487-497

    Abstract

    In Parkinson's disease (PD), dopamine depletion causes major changes in the brain, resulting in the typical cardinal motor features of the disease. PD neuropathology has been restricted to postmortem examinations, which are limited to only a single time of PD progression. Models of PD in which dopamine tone in the brain is chemically or physically disrupted are valuable tools in understanding the mechanisms of the disease. The basal ganglia have been well studied in the context of PD, and circuit changes in response to dopamine loss have been linked to the motor dysfunctions in PD. However, the etiology of the cognitive dysfunctions that are comorbid in PD patients has remained unclear until now. In this article, we review recent studies exploring how dopamine depletion affects the motor cortex at the synaptic level. In particular, we highlight our recent findings on abnormal spine dynamics in the motor cortex of PD mouse models through in vivo time-lapse imaging and motor skill behavior assays. In combination with previous studies, a role of the motor cortex in skill learning and the impairment of this ability with the loss of dopamine are becoming more apparent. Taken together, we conclude with a discussion on the potential role for the motor cortex in PD, with the possibility of targeting the motor cortex for future PD therapeutics. © 2017 International Parkinson and Movement Disorder Society.

    View details for DOI 10.1002/mds.26938

    View details for Web of Science ID 000399758800003

    View details for PubMedID 28343366

  • The Locomotion Tug-of-War: Cholinergic and Dopaminergic Interactions Outside the Striatum. Neuron Kaganovsky, K., Ding, J. B. 2017; 96 (6): 1208–10

    Abstract

    In this issue of Neuron, Moehle et al. (2017) demonstrate that presynaptic muscarinic receptors counteract the effects of dopamine in an output nucleus of the basal ganglia. They provide intracellular, anatomical, and network-level mechanisms for this cholinergic-dopaminergic interplay.

    View details for DOI 10.1016/j.neuron.2017.12.014

    View details for PubMedID 29268090

  • Selective activation of parvalbumin interneurons prevents stress-induced synapse loss and perceptual defects. Molecular psychiatry Chen, C. C., Lu, J., Yang, R., Ding, J. B., Zuo, Y. 2017

    Abstract

    Stress, a prevalent experience in modern society, is a major risk factor for many psychiatric disorders. Although sensorimotor abnormalities are often present in these disorders, little is known about how stress affects the sensory cortex. Combining behavioral analyses with in vivo synaptic imaging, we show that stressful experiences lead to progressive, clustered loss of dendritic spines along the apical dendrites of layer (L) 5 pyramidal neurons (PNs) in the mouse barrel cortex, and such spine loss closely associates with deteriorated performance in a whisker-dependent texture discrimination task. Furthermore, the activity of parvalbumin-expressing inhibitory interneurons (PV+ INs) decreases in the stressed mouse due to reduced excitability of these neurons. Importantly, both behavioral defects and structural changes of L5 PNs are prevented by selective pharmacogenetic activation of PV+INs in the barrel cortex during stress. Finally, stressed mice raised under environmental enrichment (EE) maintain normal activation of PV+ INs, normal texture discrimination, and L5 PN spine dynamics similar to unstressed EE mice. Our findings suggest that the PV+ inhibitory circuit is crucial for normal synaptic dynamics in the mouse barrel cortex and sensory function. Pharmacological, pharmacogenetic and environmental approaches to prevent stress-induced maladaptive behaviors and synaptic malfunctions converge on the regulation of PV+ IN activity, pointing to a potential therapeutic target for stress-related disorders.Molecular Psychiatry advance online publication, 1 August 2017; doi:10.1038/mp.2017.159.

    View details for DOI 10.1038/mp.2017.159

    View details for PubMedID 28761082

    View details for PubMedCentralID PMC5794672

  • TGF-beta Signaling in Dopaminergic Neurons Regulates Dendritic Growth, Excitatory-Inhibitory Synaptic Balance, and Reversal Learning CELL REPORTS Luo, S. X., Timbang, L., Kim, J., Shang, Y., Sandoval, K., Tang, A. A., Whistler, J. L., Ding, J. B., Huang, E. J. 2016; 17 (12): 3233-3245

    Abstract

    Neural circuits involving midbrain dopaminergic (DA) neurons regulate reward and goal-directed behaviors. Although local GABAergic input is known to modulate DA circuits, the mechanism that controls excitatory/inhibitory synaptic balance in DA neurons remains unclear. Here, we show that DA neurons use autocrine transforming growth factor β (TGF-β) signaling to promote the growth of axons and dendrites. Surprisingly, removing TGF-β type II receptor in DA neurons also disrupts the balance in TGF-β1 expression in DA neurons and neighboring GABAergic neurons, which increases inhibitory input, reduces excitatory synaptic input, and alters phasic firing patterns in DA neurons. Mice lacking TGF-β signaling in DA neurons are hyperactive and exhibit inflexibility in relinquishing learned behaviors and re-establishing new stimulus-reward associations. These results support a role for TGF-β in regulating the delicate balance of excitatory/inhibitory synaptic input in local microcircuits involving DA and GABAergic neurons and its potential contributions to neuropsychiatric disorders.

    View details for DOI 10.1016/j.celrep.2016.11.068

    View details for Web of Science ID 000390895600014

    View details for PubMedID 28009292

  • Live-Cell Superresolution Imaging by Pulsed STED Two-Photon Excitation Microscopy BIOPHYSICAL JOURNAL Takasaki, K. T., Ding, J. B., Sabatini, B. L. 2013; 104 (4): 770-777

    Abstract

    Two-photon laser scanning microscopy (2PLSM) allows fluorescence imaging in thick biological samples where absorption and scattering typically degrade resolution and signal collection of one-photon imaging approaches. The spatial resolution of conventional 2PLSM is limited by diffraction, and the near-infrared wavelengths used for excitation in 2PLSM preclude the accurate imaging of many small subcellular compartments of neurons. Stimulated emission depletion (STED) microscopy is a superresolution imaging modality that overcomes the resolution limit imposed by diffraction and allows fluorescence imaging of nanoscale features. Here, we describe the design and operation of a superresolution two-photon microscope using pulsed excitation and STED lasers. We examine the depth dependence of STED imaging in acute tissue slices and find enhancement of 2P resolution ranging from approximately fivefold at 20 μm to approximately twofold at 90-μm deep. The depth dependence of resolution is found to be consistent with the depth dependence of depletion efficiency, suggesting resolution is limited by STED laser propagation through turbid tissue. Finally, we achieve live imaging of dendritic spines with 60-nm resolution and demonstrate that our technique allows accurate quantification of neuronal morphology up to 30-μm deep in living brain tissue.

    View details for DOI 10.1016/j.bpj.2012.12.053

    View details for Web of Science ID 000315320200007

    View details for PubMedID 23442955

  • Dopaminergic neurons inhibit striatal output through non-canonical release of GABA NATURE Tritsch, N. X., Ding, J. B., Sabatini, B. L. 2012; 490 (7419): 262-?

    Abstract

    The substantia nigra pars compacta and ventral tegmental area contain the two largest populations of dopamine-releasing neurons in the mammalian brain. These neurons extend elaborate projections in the striatum, a large subcortical structure implicated in motor planning and reward-based learning. Phasic activation of dopaminergic neurons in response to salient or reward-predicting stimuli is thought to modulate striatal output through the release of dopamine to promote and reinforce motor action. Here we show that activation of dopamine neurons in striatal slices rapidly inhibits action potential firing in both direct- and indirect-pathway striatal projection neurons through vesicular release of the inhibitory transmitter GABA (γ-aminobutyric acid). GABA is released directly from dopaminergic axons but in a manner that is independent of the vesicular GABA transporter VGAT. Instead, GABA release requires activity of the vesicular monoamine transporter VMAT2, which is the vesicular transporter for dopamine. Furthermore, VMAT2 expression in GABAergic neurons lacking VGAT is sufficient to sustain GABA release. Thus, these findings expand the repertoire of synaptic mechanisms used by dopamine neurons to influence basal ganglia circuits, show a new substrate whose transport is dependent on VMAT2 and demonstrate that GABA can function as a bona fide co-transmitter in monoaminergic neurons.

    View details for DOI 10.1038/nature11466

    View details for Web of Science ID 000309733300050

    View details for PubMedID 23034651

  • Fasting Activation of AgRP Neurons Requires NMDA Receptors and Involves Spinogenesis and Increased Excitatory Tone NEURON Liu, T., Kong, D., Shah, B. P., Ye, C., Koda, S., Saunders, A., Ding, J. B., Yang, Z., Sabatini, B. L., Lowell, B. B. 2012; 73 (3): 511-522

    Abstract

    AgRP neuron activity drives feeding and weight gain whereas that of nearby POMC neurons does the opposite. However, the role of excitatory glutamatergic input in controlling these neurons is unknown. To address this question, we generated mice lacking NMDA receptors (NMDARs) on either AgRP or POMC neurons. Deletion of NMDARs from AgRP neurons markedly reduced weight, body fat and food intake whereas deletion from POMC neurons had no effect. Activation of AgRP neurons by fasting, as assessed by c-Fos, Agrp and Npy mRNA expression, AMPA receptor-mediated EPSCs, depolarization and firing rates, required NMDARs. Furthermore, AgRP but not POMC neurons have dendritic spines and increased glutamatergic input onto AgRP neurons caused by fasting was paralleled by an increase in spines, suggesting fasting induced synaptogenesis and spinogenesis. Thus glutamatergic synaptic transmission and its modulation by NMDARs play key roles in controlling AgRP neurons and determining the cellular and behavioral response to fasting.

    View details for DOI 10.1016/j.neuron.2011.11.027

    View details for Web of Science ID 000300140600012

    View details for PubMedID 22325203

  • Semaphorin 3E-Plexin-D1 signaling controls pathway-specific synapse formation in the striatum. Nature neuroscience Ding, J. B., Oh, W., Sabatini, B. L., Gu, C. 2012; 15 (2): 215-223

    Abstract

    The proper formation of synaptic connectivity in the mammalian brain is critical for complex behavior. In the striatum, balanced excitatory synaptic transmission from multiple sources onto two classes of principal neurons is required for coordinated and voluntary motor control. Here we show that the interaction between the secreted semaphorin 3E (Sema3E) and its receptor Plexin-D1 is a critical determinant of synaptic specificity in cortico-thalamo-striatal circuits in mice. We find that Sema3e (encoding Sema3E) is highly expressed in thalamostriatal projection neurons, whereas in the striatum Plxnd1 (encoding Plexin-D1) is selectively expressed in direct-pathway medium spiny neurons (MSNs). Despite physical intermingling of the MSNs, genetic ablation of Plxnd1 or Sema3e results in functional and anatomical rearrangement of thalamostriatal synapses specifically in direct-pathway MSNs without effects on corticostriatal synapses. Thus, our results demonstrate that Sema3E and Plexin-D1 specify the degree of glutamatergic connectivity between a specific source and target in the complex circuitry of the basal ganglia.

    View details for DOI 10.1038/nn.3003

    View details for PubMedID 22179111

    View details for PubMedCentralID PMC3267860

  • Muscarinic modulation of striatal function and circuitry. Handbook of experimental pharmacology Goldberg, J. A., Ding, J. B., Surmeier, D. J. 2012: 223-241

    Abstract

    Striatal cholinergic interneurons are pivotal modulators of the striatal circuitry involved in action selection and decision making. Although nicotinic receptors are important transducers of acetylcholine release in the striatum, muscarinic receptors are more pervasive and have been more thoroughly studied. In this review, the effects of muscarinic receptor signaling on the principal cell types in the striatum and its canonical circuits will be discussed, highlighting new insights into their role in synaptic integration and plasticity. These studies, and those that have identified new circuit elements driven by activation of nicotinic receptors, make it clear that temporally patterned activity in cholinergic interneurons must play an important role in determining the effects on striatal circuitry. These effects could be critical to the response to salient environmental stimuli that serve to direct behavior.

    View details for DOI 10.1007/978-3-642-23274-9_10

    View details for PubMedID 22222701

  • Cholinergic modulation of synaptic integration and dendritic excitability in the striatum CURRENT OPINION IN NEUROBIOLOGY Oldenburg, I. A., Ding, J. B. 2011; 21 (3): 425-432

    Abstract

    Modulatory interneurons such as, the cholinergic interneuron, are always a perplexing subject to study. Far from clear-cut distinctions such as excitatory or inhibitory, modulating interneurons can have many, often contradictory effects. The striatum is one of the most densely expressing brain areas for cholinergic markers, and actylcholine (ACh) plays an important role in regulating synaptic transmission and cellular excitability. Every cell type in the striatum has receptors for ACh. Yet even for a given cell type, ACh affecting different receptors can have seemingly opposing roles. This review highlights relevant effects of ACh on medium spiny neurons (MSNs) of the striatum and suggests how its many effects may work in concert to modulate MSN firing properties.

    View details for DOI 10.1016/j.conb.2011.04.004

    View details for Web of Science ID 000294097100010

    View details for PubMedID 21550798

  • Thalamic Gating of Corticostriatal Signaling by Cholinergic Interneurons NEURON Ding, J. B., Guzman, J. N., Peterson, J. D., Goldberg, J. A., Surmeier, D. J. 2010; 67 (2): 294-307

    Abstract

    Salient stimuli redirect attention and suppress ongoing motor activity. This attentional shift is thought to rely upon thalamic signals to the striatum to shift cortically driven action selection, but the network mechanisms underlying this interaction are unclear. Using a brain slice preparation that preserved cortico- and thalamostriatal connectivity, it was found that activation of thalamostriatal axons in a way that mimicked the response to salient stimuli induced a burst of spikes in striatal cholinergic interneurons that was followed by a pause lasting more than half a second. This patterned interneuron activity triggered a transient, presynaptic suppression of cortical input to both major classes of principal medium spiny neuron (MSN) that gave way to a prolonged enhancement of postsynaptic responsiveness in striatopallidal MSNs controlling motor suppression. This differential regulation of the corticostriatal circuitry provides a neural substrate for attentional shifts and cessation of ongoing motor activity with the appearance of salient environmental stimuli.

    View details for DOI 10.1016/j.neuron.2010.06.017

    View details for Web of Science ID 000280461500013

    View details for PubMedID 20670836

  • Supraresolution Imaging in Brain Slices using Stimulated-Emission Depletion Two-Photon Laser Scanning Microscopy NEURON Ding, J. B., Takasaki, K. T., Sabatini, B. L. 2009; 63 (4): 429-437

    Abstract

    Two-photon laser scanning microscopy (2PLSM) has allowed unprecedented fluorescence imaging of neuronal structure and function within neural tissue. However, the resolution of this approach is poor compared to that of conventional confocal microscopy. Here, we demonstrate supraresolution 2PLSM within brain slices. Imaging beyond the diffraction limit is accomplished by using near-infrared (NIR) lasers for both pulsed two-photon excitation and continuous wave stimulated emission depletion (STED). Furthermore, we demonstrate that Alexa Fluor 594, a bright fluorophore commonly used for both live cell and fixed tissue fluorescence imaging, is suitable for STED 2PLSM. STED 2PLSM supraresolution microscopy achieves approximately 3-fold improvement in resolution in the radial direction over conventional 2PLSM, revealing greater detail in the structure of dendritic spines located approximately 100 microns below the surface of brain slices. Further improvements in resolution are theoretically achievable, suggesting that STED 2PLSM will permit nanoscale imaging of neuronal structures located in relatively intact brain tissue.

    View details for DOI 10.1016/j.neuron.2009.07.011

    View details for Web of Science ID 000269570400004

    View details for PubMedID 19709626

  • Corticostriatal and thalamostriatal synapses have distinctive properties JOURNAL OF NEUROSCIENCE Ding, J., Peterson, J. D., Surmeier, D. J. 2008; 28 (25): 6483-6492

    Abstract

    The two principal excitatory glutamatergic inputs to striatal medium spiny neurons (MSNs) arise from neurons in the cerebral cortex and thalamus. Although there have been many electrophysiological studies of MSN glutamatergic synapses, little is known about how corticostriatal and thalamostriatal synapses differ. Using mouse brain slices that allowed each type of synapse to be selectively activated, electrophysiological approaches were used to characterize their properties in identified striatopallidal and striatonigral MSNs. At corticostriatal synapses, a single afferent volley increased the glutamate released by a subsequent volley, leading to enhanced postsynaptic depolarization with repetitive stimulation. This was true for both striatonigral and striatopallidal MSNs. In contrast, at thalamostriatal synapses, a single afferent volley decreased glutamate released by a subsequent volley, leading to a depressed postsynaptic depolarization with repetitive stimulation. Again, this response pattern was the same in striatonigral and striatopallidal MSNs. These differences in release probability and short-term synaptic plasticity suggest that corticostriatal and thalamostriatal projection systems code information in temporally distinct ways, constraining how they regulate striatal circuitry.

    View details for DOI 10.1523/JNEUROSCI.0435-08.2008

    View details for Web of Science ID 000256890000022

    View details for PubMedID 18562619

  • Re-emergence of striatal cholinergic interneurons in movement disorders TRENDS IN NEUROSCIENCES Pisani, A., Bernardi, G., Ding, J., Surmeier, D. J. 2007; 30 (10): 545-553

    Abstract

    Twenty years ago, striatal cholinergic neurons were central figures in models of basal ganglia function. But since then, they have receded in importance. Recent studies are likely to lead to their re-emergence in our thinking. Cholinergic interneurons have been implicated as key players in the induction of synaptic plasticity and motor learning, as well as in motor dysfunction. In Parkinson's disease and dystonia, diminished striatal dopaminergic signalling leads to increased release of acetylcholine by interneurons, distorting network function and inducing structural changes that undoubtedly contribute to the symptoms. By contrast, in Huntington's disease and progressive supranuclear palsy, there is a fall in striatal cholinergic markers. This review gives an overview of these recent experimental and clinical studies, placing them within the context of the pathogenesis of movement disorders.

    View details for DOI 10.1016/j.tins.2007.07.008

    View details for Web of Science ID 000250679500009

    View details for PubMedID 17904652

  • D1 and D2 dopamine-receptor modulation of striatal glutamatergic signaling in striatal medium spiny neurons TRENDS IN NEUROSCIENCES Surmeier, D. J., Ding, J., Day, M., Wang, Z., Shen, W. 2007; 30 (5): 228-235

    Abstract

    Dopamine shapes a wide variety of psychomotor functions. This is mainly accomplished by modulating cortical and thalamic glutamatergic signals impinging upon principal medium spiny neurons (MSNs) of the striatum. Several lines of evidence suggest that dopamine D1 receptor signaling enhances dendritic excitability and glutamatergic signaling in striatonigral MSNs, whereas D2 receptor signaling exerts the opposite effect in striatopallidal MSNs. The functional antagonism between these two major striatal dopamine receptors extends to the regulation of synaptic plasticity. Recent studies, using transgenic mice in which cells express D1 and D2 receptors, have uncovered unappreciated differences between MSNs that shape glutamatergic signaling and the influence of DA on synaptic plasticity. These studies have also shown that long-term alterations in dopamine signaling produce profound and cell-type-specific reshaping of corticostriatal connectivity and function.

    View details for DOI 10.1016/j.tins.2007.03.008

    View details for Web of Science ID 000246752000007

    View details for PubMedID 17408758

  • RGS4-dependent attenuation of M-4 autoreceptor function in striatal cholinergic interneurons following dopamine depletion NATURE NEUROSCIENCE Ding, J., Guzman, J. N., Tkatch, T., chen, s., Goldberg, J. A., Ebert, P. J., Levitt, P., Wilson, C. J., Hamm, H. E., Surmeier, D. J. 2006; 9 (6): 832-842

    Abstract

    Parkinson disease is a neurodegenerative disorder whose symptoms are caused by the loss of dopaminergic neurons innervating the striatum. As striatal dopamine levels fall, striatal acetylcholine release rises, exacerbating motor symptoms. This adaptation is commonly attributed to the loss of interneuronal regulation by inhibitory D(2) dopamine receptors. Our results point to a completely different, new mechanism. After striatal dopamine depletion, D(2) dopamine receptor modulation of calcium (Ca(2+)) channels controlling vesicular acetylcholine release in interneurons was unchanged, but M(4) muscarinic autoreceptor coupling to these same channels was markedly attenuated. This adaptation was attributable to the upregulation of RGS4-an autoreceptor-associated, GTPase-accelerating protein. This specific signaling adaptation extended to a broader loss of autoreceptor control of interneuron spiking. These observations suggest that RGS4-dependent attenuation of interneuronal autoreceptor signaling is a major factor in the elevation of striatal acetylcholine release in Parkinson disease.

    View details for DOI 10.1038/nn1700

    View details for Web of Science ID 000237895200024

    View details for PubMedID 16699510

  • Dopaminergic control of corticostriatal long-term synaptic depression in medium spiny neurons is mediated by cholinergic interneurons NEURON Wang, Z., Kai, L., Day, M., Ronesi, J., Yin, H. H., Ding, J., Tkatch, T., Lovinger, D. M., Surmeier, D. J. 2006; 50 (3): 443-452

    Abstract

    Long-term depression (LTD) of the synapse formed between cortical pyramidal neurons and striatal medium spiny neurons is central to many theories of motor plasticity and associative learning. The induction of LTD at this synapse is thought to depend upon D(2) dopamine receptors localized in the postsynaptic membrane. If this were true, LTD should be inducible in neurons from only one of the two projection systems of the striatum. Using transgenic mice in which neurons that contribute to these two systems are labeled, we show that this is not the case. Rather, in both cell types, the D(2) receptor dependence of LTD induction reflects the need to lower M(1) muscarinic receptor activity-a goal accomplished by D(2) receptors on cholinergic interneurons. In addition to reconciling discordant tracts of the striatal literature, these findings point to cholinergic interneurons as key mediators of dopamine-dependent striatal plasticity and learning.

    View details for DOI 10.1016/j.neuron.2006.04.010

    View details for Web of Science ID 000237726800013

    View details for PubMedID 16675398

  • Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models NATURE NEUROSCIENCE Day, M., Wang, Z. F., Ding, J., An, X. H., Ingham, C. A., Shering, A. F., Wokosin, D., Ilijic, E., Sun, Z. X., Sampson, A. R., Mugnaini, E., Deutch, A. Y., Sesack, S. R., Arbuthnott, G. W., Surmeier, D. J. 2006; 9 (2): 251-259

    Abstract

    Parkinson disease is a common neurodegenerative disorder that leads to difficulty in effectively translating thought into action. Although it is known that dopaminergic neurons that innervate the striatum die in Parkinson disease, it is not clear how this loss leads to symptoms. Recent work has implicated striatopallidal medium spiny neurons (MSNs) in this process, but how and precisely why these neurons change is not clear. Using multiphoton imaging, we show that dopamine depletion leads to a rapid and profound loss of spines and glutamatergic synapses on striatopallidal MSNs but not on neighboring striatonigral MSNs. This loss of connectivity is triggered by a new mechanism-dysregulation of intraspine Cav1.3 L-type Ca(2+) channels. The disconnection of striatopallidal neurons from motor command structures is likely to be a key step in the emergence of pathological activity that is responsible for symptoms in Parkinson disease.

    View details for DOI 10.1038/nn1632

    View details for Web of Science ID 000234990100021

    View details for PubMedID 16415865