Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.691
Filter
1.
Cell ; 187(14): 3671-3689.e23, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38866017

ABSTRACT

Ongoing, early-stage clinical trials illustrate the translational potential of human pluripotent stem cell (hPSC)-based cell therapies in Parkinson's disease (PD). However, an unresolved challenge is the extensive cell death following transplantation. Here, we performed a pooled CRISPR-Cas9 screen to enhance postmitotic dopamine neuron survival in vivo. We identified p53-mediated apoptotic cell death as a major contributor to dopamine neuron loss and uncovered a causal link of tumor necrosis factor alpha (TNF-α)-nuclear factor κB (NF-κB) signaling in limiting cell survival. As a translationally relevant strategy to purify postmitotic dopamine neurons, we identified cell surface markers that enable purification without the need for genetic reporters. Combining cell sorting and treatment with adalimumab, a clinically approved TNF-α inhibitor, enabled efficient engraftment of postmitotic dopamine neurons with extensive reinnervation and functional recovery in a preclinical PD mouse model. Thus, transient TNF-α inhibition presents a clinically relevant strategy to enhance survival and enable engraftment of postmitotic hPSC-derived dopamine neurons in PD.


Subject(s)
Cell Survival , Dopaminergic Neurons , NF-kappa B , Tumor Necrosis Factor-alpha , Tumor Suppressor Protein p53 , Dopaminergic Neurons/metabolism , Animals , Humans , NF-kappa B/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Necrosis Factor-alpha/metabolism , Mice , Cell Survival/drug effects , Signal Transduction , Parkinson Disease/metabolism , Pluripotent Stem Cells/metabolism , Apoptosis , Disease Models, Animal , CRISPR-Cas Systems
2.
Cell ; 186(3): 577-590.e16, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36693373

ABSTRACT

Pleasurable touch is paramount during social behavior, including sexual encounters. However, the identity and precise role of sensory neurons that transduce sexual touch remain unknown. A population of sensory neurons labeled by developmental expression of the G protein-coupled receptor Mrgprb4 detects mechanical stimulation in mice. Here, we study the social relevance of Mrgprb4-lineage neurons and reveal that these neurons are required for sexual receptivity and sufficient to induce dopamine release in the brain. Even in social isolation, optogenetic stimulation of Mrgprb4-lineage neurons through the back skin is sufficient to induce a conditioned place preference and a striking dorsiflexion resembling the lordotic copulatory posture. In the absence of Mrgprb4-lineage neurons, female mice no longer find male mounts rewarding: sexual receptivity is supplanted by aggression and a coincident decline in dopamine release in the nucleus accumbens. Together, these findings establish that Mrgprb4-lineage neurons initiate a skin-to-brain circuit encoding the rewarding quality of social touch.


Subject(s)
Dopamine , Touch , Mice , Male , Female , Animals , Dopamine/metabolism , Nucleus Accumbens/metabolism , Sensory Receptor Cells/metabolism , Skin/metabolism , Reward , Dopaminergic Neurons/metabolism , Optogenetics , Receptors, G-Protein-Coupled/metabolism
3.
Cell ; 185(11): 1943-1959.e21, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35545089

ABSTRACT

Parthanatos-associated apoptosis-inducing factor (AIF) nuclease (PAAN), also known as macrophage migration inhibitor factor (MIF), is a member of the PD-D/E(X)K nucleases that acts as a final executioner in parthanatos. PAAN's role in Parkinson's disease (PD) and whether it is amenable to chemical inhibition is not known. Here, we show that neurodegeneration induced by pathologic α-synuclein (α-syn) occurs via PAAN/MIF nuclease activity. Genetic depletion of PAAN/MIF and a mutant lacking nuclease activity prevent the loss of dopaminergic neurons and behavioral deficits in the α-syn preformed fibril (PFF) mouse model of sporadic PD. Compound screening led to the identification of PAANIB-1, a brain-penetrant PAAN/MIF nuclease inhibitor that prevents neurodegeneration induced by α-syn PFF, AAV-α-syn overexpression, or MPTP intoxication in vivo. Our findings could have broad relevance in human pathologies where parthanatos plays a role in the development of cell death inhibitors targeting the druggable PAAN/MIF nuclease.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Parkinson Disease , Animals , Brain/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Endonucleases/metabolism , Mice , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism
4.
Cell ; 178(1): 60-75.e19, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31230716

ABSTRACT

Animals rely on the relative timing of events in their environment to form and update predictive associations, but the molecular and circuit mechanisms for this temporal sensitivity remain incompletely understood. Here, we show that olfactory associations in Drosophila can be written and reversed on a trial-by-trial basis depending on the temporal relationship between an odor cue and dopaminergic reinforcement. Through the synchronous recording of neural activity and behavior, we show that reversals in learned odor attraction correlate with bidirectional neural plasticity in the mushroom body, the associative olfactory center of the fly. Two dopamine receptors, DopR1 and DopR2, contribute to this temporal sensitivity by coupling to distinct second messengers and directing either synaptic depression or potentiation. Our results reveal how dopamine-receptor signaling pathways can detect the order of events to instruct opposing forms of synaptic and behavioral plasticity, allowing animals to flexibly update their associations in a dynamic environment.


Subject(s)
Association Learning/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Mushroom Bodies/physiology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine/metabolism , Animals , Behavior, Animal/physiology , Conditioning, Classical/physiology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Neuronal Plasticity , Odorants , Reward , Smell/physiology , Synaptic Potentials/physiology , Time Factors
5.
Cell ; 175(3): 709-722.e15, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30245010

ABSTRACT

Accurately predicting an outcome requires that animals learn supporting and conflicting evidence from sequential experience. In mammals and invertebrates, learned fear responses can be suppressed by experiencing predictive cues without punishment, a process called memory extinction. Here, we show that extinction of aversive memories in Drosophila requires specific dopaminergic neurons, which indicate that omission of punishment is remembered as a positive experience. Functional imaging revealed co-existence of intracellular calcium traces in different places in the mushroom body output neuron network for both the original aversive memory and a new appetitive extinction memory. Light and ultrastructural anatomy are consistent with parallel competing memories being combined within mushroom body output neurons that direct avoidance. Indeed, extinction-evoked plasticity in a pair of these neurons neutralizes the potentiated odor response imposed in the network by aversive learning. Therefore, flies track the accuracy of learned expectations by accumulating and integrating memories of conflicting events.


Subject(s)
Extinction, Psychological , Memory , Animals , Appetitive Behavior , Calcium/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Drosophila melanogaster , Female , Mushroom Bodies/cytology , Mushroom Bodies/physiology , Neuronal Plasticity
6.
Cell ; 174(6): 1436-1449.e20, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30146163

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.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Dopaminergic Neurons/metabolism , Nuclear Proteins/genetics , Synapses/metabolism , Active Transport, Cell Nucleus , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Calcium Signaling , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis , Mutation, Missense , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/metabolism
7.
Cell ; 166(6): 1564-1571.e6, 2016 Sep 08.
Article in English | MEDLINE | ID: mdl-27610576

ABSTRACT

Optogenetic studies in mice have revealed new relationships between well-defined neurons and brain functions. However, there are currently no means to achieve the same cell-type specificity in monkeys, which possess an expanded behavioral repertoire and closer anatomical homology to humans. Here, we present a resource for cell-type-specific channelrhodopsin expression in Rhesus monkeys and apply this technique to modulate dopamine activity and monkey choice behavior. These data show that two viral vectors label dopamine neurons with greater than 95% specificity. Infected neurons were activated by light pulses, indicating functional expression. The addition of optical stimulation to reward outcomes promoted the learning of reward-predicting stimuli at the neuronal and behavioral level. Together, these results demonstrate the feasibility of effective and selective stimulation of dopamine neurons in non-human primates and a resource that could be applied to other cell types in the monkey brain.


Subject(s)
Choice Behavior/physiology , Dopaminergic Neurons/metabolism , Optogenetics/methods , Animals , Dependovirus/genetics , Dopamine/metabolism , Gene Expression Regulation , Genetic Vectors/genetics , Macaca mulatta , Promoter Regions, Genetic/genetics , Rhodopsin/genetics
8.
Nature ; 626(7999): 583-592, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38092040

ABSTRACT

Animals exhibit a diverse behavioural repertoire when exploring new environments and can learn which actions or action sequences produce positive outcomes. Dopamine release after encountering a reward is critical for reinforcing reward-producing actions1-3. However, it has been challenging to understand how credit is assigned to the exact action that produced the dopamine release during continuous behaviour. Here we investigated this problem in mice using a self-stimulation paradigm in which specific spontaneous movements triggered optogenetic stimulation of dopaminergic neurons. Dopamine self-stimulation rapidly and dynamically changes the structure of the entire behavioural repertoire. Initial stimulations reinforced not only the stimulation-producing target action, but also actions similar to the target action and actions that occurred a few seconds before stimulation. Repeated pairings led to a gradual refinement of the behavioural repertoire to home in on the target action. Reinforcement of action sequences revealed further temporal dependencies of refinement. Action pairs spontaneously separated by long time intervals promoted a stepwise credit assignment, with early refinement of actions most proximal to stimulation and subsequent refinement of more distal actions. Thus, a retrospective reinforcement mechanism promotes not only reinforcement, but also gradual refinement of the entire behavioural repertoire to assign credit to specific actions and action sequences that lead to dopamine release.


Subject(s)
Dopamine , Learning , Reinforcement, Psychology , Reward , Animals , Mice , Decision Making/physiology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Learning/physiology , Optogenetics , Time Factors , Models, Psychological , Models, Neurological
9.
Nature ; 630(8015): 141-148, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38778097

ABSTRACT

Fentanyl is a powerful painkiller that elicits euphoria and positive reinforcement1. Fentanyl also leads to dependence, defined by the aversive withdrawal syndrome, which fuels negative reinforcement2,3 (that is, individuals retake the drug to avoid withdrawal). Positive and negative reinforcement maintain opioid consumption, which leads to addiction in one-fourth of users, the largest fraction for all addictive drugs4. Among the opioid receptors, µ-opioid receptors have a key role5, yet the induction loci of circuit adaptations that eventually lead to addiction remain unknown. Here we injected mice with fentanyl to acutely inhibit γ-aminobutyric acid-expressing neurons in the ventral tegmental area (VTA), causing disinhibition of dopamine neurons, which eventually increased dopamine in the nucleus accumbens. Knockdown of µ-opioid receptors in VTA abolished dopamine transients and positive reinforcement, but withdrawal remained unchanged. We identified neurons expressing µ-opioid receptors in the central amygdala (CeA) whose activity was enhanced during withdrawal. Knockdown of µ-opioid receptors in CeA eliminated aversive symptoms, suggesting that they mediate negative reinforcement. Thus, optogenetic stimulation caused place aversion, and mice readily learned to press a lever to pause optogenetic stimulation of CeA neurons that express µ-opioid receptors. Our study parses the neuronal populations that trigger positive and negative reinforcement in VTA and CeA, respectively. We lay out the circuit organization to develop interventions for reducing fentanyl addiction and facilitating rehabilitation.


Subject(s)
Fentanyl , Receptors, Opioid, mu , Reinforcement, Psychology , Animals , Female , Male , Mice , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage , Central Amygdaloid Nucleus/cytology , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/metabolism , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Fentanyl/pharmacology , Mice, Inbred C57BL , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/pathology , Optogenetics , Receptors, Opioid, mu/metabolism , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
10.
Nature ; 630(8017): 677-685, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839962

ABSTRACT

All drugs of abuse induce long-lasting changes in synaptic transmission and neural circuit function that underlie substance-use disorders1,2. Another recently appreciated mechanism of neural circuit plasticity is mediated through activity-regulated changes in myelin that can tune circuit function and influence cognitive behaviour3-7. Here we explore the role of myelin plasticity in dopaminergic circuitry and reward learning. We demonstrate that dopaminergic neuronal activity-regulated myelin plasticity is a key modulator of dopaminergic circuit function and opioid reward. Oligodendroglial lineage cells respond to dopaminergic neuronal activity evoked by optogenetic stimulation of dopaminergic neurons, optogenetic inhibition of GABAergic neurons, or administration of morphine. These oligodendroglial changes are evident selectively within the ventral tegmental area but not along the axonal projections in the medial forebrain bundle nor within the target nucleus accumbens. Genetic blockade of oligodendrogenesis dampens dopamine release dynamics in nucleus accumbens and impairs behavioural conditioning to morphine. Taken together, these findings underscore a critical role for oligodendrogenesis in reward learning and identify dopaminergic neuronal activity-regulated myelin plasticity as an important circuit modification that is required for opioid reward.


Subject(s)
Analgesics, Opioid , Myelin Sheath , Neural Pathways , Neuronal Plasticity , Reward , Ventral Tegmental Area , Animals , Female , Male , Mice , Analgesics, Opioid/pharmacology , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Mice, Inbred C57BL , Morphine/pharmacology , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Nucleus Accumbens/drug effects , Oligodendroglia/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , Optogenetics , Ventral Tegmental Area/physiology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Neural Pathways/drug effects , Cell Lineage
11.
Nat Rev Neurosci ; 25(6): 393-413, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38600347

ABSTRACT

Parkinson disease (PD) is a neurodegenerative disorder marked by the preferential dysfunction and death of dopaminergic neurons in the substantia nigra. The onset and progression of PD is influenced by a diversity of genetic variants, many of which lack functional characterization. To identify the most high-yield targets for therapeutic intervention, it is important to consider the core cellular compartments and functional pathways upon which the varied forms of pathogenic dysfunction may converge. Here, we review several key PD-linked proteins and pathways, focusing on the mechanisms of their potential convergence in disease pathogenesis. These dysfunctions primarily localize to a subset of subcellular compartments, including mitochondria, lysosomes and synapses. We discuss how these pathogenic mechanisms that originate in different cellular compartments may coordinately lead to cellular dysfunction and neurodegeneration in PD.


Subject(s)
Parkinson Disease , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease/metabolism , Humans , Animals , Mitochondria/genetics , Mitochondria/metabolism , Dopaminergic Neurons/pathology , Dopaminergic Neurons/metabolism , Lysosomes/metabolism , Lysosomes/genetics , Synapses/pathology , Synapses/genetics , Synapses/metabolism
12.
Nature ; 616(7957): 510-519, 2023 04.
Article in English | MEDLINE | ID: mdl-37020025

ABSTRACT

The central amygdala (CeA) is implicated in a range of mental processes including attention, motivation, memory formation and extinction and in behaviours driven by either aversive or appetitive stimuli1-7. How it participates in these divergent functions remains elusive. Here we show that somatostatin-expressing (Sst+) CeA neurons, which mediate much of CeA functions3,6,8-10, generate experience-dependent and stimulus-specific evaluative signals essential for learning. The population responses of these neurons in mice encode the identities of a wide range of salient stimuli, with the responses of separate subpopulations selectively representing the stimuli that have contrasting valences, sensory modalities or physical properties (for example, shock and water reward). These signals scale with stimulus intensity, undergo pronounced amplification and transformation during learning, and are required for both reward and aversive learning. Notably, these signals contribute to the responses of dopamine neurons to reward and reward prediction error, but not to their responses to aversive stimuli. In line with this, Sst+ CeA neuron outputs to dopamine areas are required for reward learning, but are dispensable for aversive learning. Our results suggest that Sst+ CeA neurons selectively process information about differing salient events for evaluation during learning, supporting the diverse roles of the CeA. In particular, the information for dopamine neurons facilitates reward evaluation.


Subject(s)
Avoidance Learning , Central Amygdaloid Nucleus , Neuronal Plasticity , Reward , Animals , Mice , Avoidance Learning/physiology , Central Amygdaloid Nucleus/cytology , Central Amygdaloid Nucleus/physiology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Motivation , Somatostatin/metabolism , Electroshock
13.
Nature ; 621(7979): 543-549, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37558873

ABSTRACT

External rewards such as food and money are potent modifiers of behaviour1,2. Pioneering studies established that these salient sensory stimuli briefly interrupt the tonic discharge of neurons that produce the neuromodulators dopamine (DA) and acetylcholine (ACh): midbrain DA neurons (DANs) fire a burst of action potentials that broadly elevates DA in the striatum3,4 at the same time that striatal cholinergic interneurons (CINs) produce a characteristic pause in firing5,6. These phasic responses are thought to create unique, temporally limited conditions that motivate action and promote learning7-11. However, the dynamics of DA and ACh outside explicitly rewarded situations remain poorly understood. Here we show that extracellular DA and ACh levels fluctuate spontaneously and periodically at a frequency of approximately 2 Hz in the dorsal striatum of mice and maintain the same temporal relationship relative to one another as that evoked by reward. We show that this neuromodulatory coordination does not arise from direct interactions between DA and ACh within the striatum. Instead, we provide evidence that periodic fluctuations in striatal DA are inherited from midbrain DANs, while striatal ACh transients are driven by glutamatergic inputs, which act to locally synchronize the spiking of CINs. Together, our findings show that striatal neuromodulatory dynamics are autonomously organized by distributed extra-striatal afferents. The dominance of intrinsic rhythms in DA and ACh offers new insights for explaining how reward-associated neural dynamics emerge and how the brain motivates action and promotes learning from within.


Subject(s)
Acetylcholine , Corpus Striatum , Dopamine , Animals , Mice , Acetylcholine/metabolism , Action Potentials , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Glutamine/metabolism , Interneurons/metabolism , Motivation , Neostriatum/cytology , Neostriatum/metabolism , Reward , Afferent Pathways
14.
Nature ; 623(7986): 375-380, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37758948

ABSTRACT

Hunger, thirst, loneliness and ambition determine the reward value of food, water, social interaction and performance outcome1. Dopamine neurons respond to rewards meeting these diverse needs2-8, but it remains unclear how behaviour and dopamine signals change as priorities change with new opportunities in the environment. One possibility is that dopamine signals for distinct drives are routed to distinct dopamine pathways9,10. Another possibility is that dopamine signals in a given pathway are dynamically tuned to rewards set by the current priority. Here we used electrophysiology and fibre photometry to test how dopamine signals associated with quenching thirst, singing a good song and courting a mate change as male zebra finches (Taeniopygia guttata) were provided with opportunities to retrieve water, evaluate song performance or court a female. When alone, water reward signals were observed in two mesostriatal pathways but singing-related performance error signals were routed to Area X, a striatal nucleus specialized for singing. When courting a female, water seeking was reduced and dopamine responses to both water and song performance outcomes diminished. Instead, dopamine signals in Area X were driven by female calls timed with the courtship song. Thus the dopamine system handled coexisting drives by routing vocal performance and social feedback signals to a striatal area for communication and by flexibly re-tuning to rewards set by the prioritized drive.


Subject(s)
Brain , Courtship , Dopamine , Dopaminergic Neurons , Feedback, Physiological , Feedback, Psychological , Finches , Animals , Female , Male , Dopamine/metabolism , Finches/physiology , Vocalization, Animal/physiology , Water , Feedback, Physiological/physiology , Drinking/physiology , Thirst/physiology , Dopaminergic Neurons/metabolism , Electrophysiology , Brain/cytology , Brain/physiology , Communication , Reward , Feedback, Psychological/physiology
15.
Nature ; 617(7962): 777-784, 2023 May.
Article in English | MEDLINE | ID: mdl-37100911

ABSTRACT

Associating multiple sensory cues with objects and experience is a fundamental brain process that improves object recognition and memory performance. However, neural mechanisms that bind sensory features during learning and augment memory expression are unknown. Here we demonstrate multisensory appetitive and aversive memory in Drosophila. Combining colours and odours improved memory performance, even when each sensory modality was tested alone. Temporal control of neuronal function revealed visually selective mushroom body Kenyon cells (KCs) to be required for enhancement of both visual and olfactory memory after multisensory training. Voltage imaging in head-fixed flies showed that multisensory learning binds activity between streams of modality-specific KCs so that unimodal sensory input generates a multimodal neuronal response. Binding occurs between regions of the olfactory and visual KC axons, which receive valence-relevant dopaminergic reinforcement, and is propagated downstream. Dopamine locally releases GABAergic inhibition to permit specific microcircuits within KC-spanning serotonergic neurons to function as an excitatory bridge between the previously 'modality-selective' KC streams. Cross-modal binding thereby expands the KCs representing the memory engram for each modality into those representing the other. This broadening of the engram improves memory performance after multisensory learning and permits a single sensory feature to retrieve the memory of the multimodal experience.


Subject(s)
Brain , Color Perception , Drosophila melanogaster , Learning , Memory , Neurons , Olfactory Perception , Animals , Brain/cytology , Brain/physiology , Dopamine/metabolism , Learning/physiology , Mushroom Bodies/cytology , Mushroom Bodies/physiology , Neurons/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , GABAergic Neurons/metabolism , Serotonergic Neurons/metabolism , Memory/physiology , Olfactory Perception/physiology , Dopaminergic Neurons/metabolism , Neural Inhibition , Color Perception/physiology , Odorants/analysis
16.
Nature ; 619(7969): 332-337, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380765

ABSTRACT

Fast-acting neurotransmitters and slow, modulatory neuropeptides are co-released from neurons in the central nervous system, albeit from distinct synaptic vesicles1. The mechanisms of how co-released neurotransmitters and neuropeptides that have opposing actions-for example, stimulatory versus inhibitory-work together to exert control of neural circuit output remain unclear. This has been difficult to resolve owing to the inability to selectively isolate these signalling pathways in a cell- and circuit-specific manner. Here we developed a genetic-based anatomical disconnect procedure that utilizes distinct DNA recombinases to independently facilitate CRISPR-Cas9 mutagenesis2 of neurotransmitter- and neuropeptide-related genes in distinct cell types in two different brain regions simultaneously. We demonstrate that neurons within the lateral hypothalamus that produce the stimulatory neuropeptide neurotensin and the inhibitory neurotransmitter GABA (γ-aminobutyric acid) utilize these signals to coordinately activate dopamine-producing neurons of the ventral tegmental area. We show that GABA release from lateral hypothalamus neurotensin neurons inhibits GABA neurons within the ventral tegmental area, disinhibiting dopamine neurons and causing a rapid rise in calcium, whereas neurotensin directly generates a slow inactivating calcium signal in dopamine neurons that is dependent on the expression of neurotensin receptor 1 (Ntsr1). We further show that these two signals work together to regulate dopamine neuron responses to maximize behavioural responding. Thus, a neurotransmitter and a neuropeptide with opposing signals can act on distinct timescales through different cell types to enhance circuit output and optimize behaviour.


Subject(s)
Brain , Neural Pathways , Neurotensin , Neurotransmitter Agents , Signal Transduction , Brain/cytology , Brain/metabolism , Calcium/metabolism , CRISPR-Cas Systems , Dopamine/metabolism , Dopaminergic Neurons/metabolism , GABAergic Neurons , gamma-Aminobutyric Acid/metabolism , Gene Editing , Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/metabolism , Neurotensin/metabolism , Neurotransmitter Agents/metabolism , Receptors, Neurotensin/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/metabolism
17.
Nature ; 619(7970): 606-615, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37438521

ABSTRACT

The specific loss of midbrain dopamine neurons (mDANs) causes major motor dysfunction in Parkinson's disease, which makes cell replacement a promising therapeutic approach1-4. However, poor survival of grafted mDANs remains an obstacle to successful clinical outcomes5-8. Here we show that the surgical procedure itself (referred to here as 'needle trauma') triggers a profound host response that is characterized by acute neuroinflammation, robust infiltration of peripheral immune cells and brain cell death. When midbrain dopamine (mDA) cells derived from human induced pluripotent stem (iPS) cells were transplanted into the rodent striatum, less than 10% of implanted tyrosine hydroxylase (TH)+ mDANs survived at two weeks after transplantation. By contrast, TH- grafted cells mostly survived. Notably, transplantation of autologous regulatory T (Treg) cells greatly modified the response to needle trauma, suppressing acute neuroinflammation and immune cell infiltration. Furthermore, intra-striatal co-transplantation of Treg cells and human-iPS-cell-derived mDA cells significantly protected grafted mDANs from needle-trauma-associated death and improved therapeutic outcomes in rodent models of Parkinson's disease with 6-hydroxydopamine lesions. Co-transplantation with Treg cells also suppressed the undesirable proliferation of TH- grafted cells, resulting in more compact grafts with a higher proportion and higher absolute numbers of TH+ neurons. Together, these data emphasize the importance of the initial inflammatory response to surgical injury in the differential survival of cellular components of the graft, and suggest that co-transplanting autologous Treg cells effectively reduces the needle-trauma-induced death of mDANs, providing a potential strategy to achieve better clinical outcomes for cell therapy in Parkinson's disease.


Subject(s)
Cell- and Tissue-Based Therapy , Dopaminergic Neurons , Graft Survival , Neuroinflammatory Diseases , Parkinson Disease , T-Lymphocytes, Regulatory , Tyrosine 3-Monooxygenase , Humans , Dopamine/analogs & derivatives , Dopamine/metabolism , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/transplantation , Mesencephalon/pathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/prevention & control , Neuroinflammatory Diseases/therapy , Parkinson Disease/complications , Parkinson Disease/pathology , Parkinson Disease/surgery , Parkinson Disease/therapy , Tyrosine 3-Monooxygenase/deficiency , Tyrosine 3-Monooxygenase/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Cell- and Tissue-Based Therapy/methods , Animals , Mice , Rats , Oxidopamine/metabolism , Graft Survival/immunology , Cell Death , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Neostriatum/metabolism , Time Factors , Cell Proliferation , Treatment Outcome
18.
Cell ; 155(4): 881-93, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209625

ABSTRACT

Behavioral persistence is a major factor in determining when and under which circumstances animals will terminate their current activity and transition into more profitable, appropriate, or urgent behavior. We show that, for the first 5 min of copulation in Drosophila, stressful stimuli do not interrupt mating, whereas 10 min later, even minor perturbations are sufficient to terminate copulation. This decline in persistence occurs as the probability of successful mating increases and is promoted by approximately eight sexually dimorphic, GABAergic interneurons of the male abdominal ganglion. When these interneurons were silenced, persistence increased and males copulated far longer than required for successful mating. When these interneurons were stimulated, persistence decreased and copulations were shortened. In contrast, dopaminergic neurons of the ventral nerve cord promote copulation persistence and extend copulation duration. Thus, copulation duration in Drosophila is a product of gradually declining persistence controlled by opposing neuronal populations using conserved neurotransmission systems.


Subject(s)
Dopaminergic Neurons/metabolism , Drosophila melanogaster/physiology , GABAergic Neurons/metabolism , Sexual Behavior, Animal , Animals , Copulation , Drosophila melanogaster/cytology , Female , Male
19.
Nature ; 608(7922): 368-373, 2022 08.
Article in English | MEDLINE | ID: mdl-35896744

ABSTRACT

Ketamine is used clinically as an anaesthetic and a fast-acting antidepressant, and recreationally for its dissociative properties, raising concerns of addiction as a possible side effect. Addictive drugs such as cocaine increase the levels of dopamine in the nucleus accumbens. This facilitates synaptic plasticity in the mesolimbic system, which causes behavioural adaptations and eventually drives the transition to compulsion1-4. The addiction liability of ketamine is a matter of much debate, in part because of its complex pharmacology that among several targets includes N-methyl-D-aspartic acid (NMDA) receptor (NMDAR) antagonism5,6. Here we show that ketamine does not induce the synaptic plasticity that is typically observed with addictive drugs in mice, despite eliciting robust dopamine transients in the nucleus accumbens. Ketamine nevertheless supported reinforcement through the disinhibition of dopamine neurons in the ventral tegmental area (VTA). This effect was mediated by NMDAR antagonism in GABA (γ-aminobutyric acid) neurons of the VTA, but was quickly terminated by type-2 dopamine receptors on dopamine neurons. The rapid off-kinetics of the dopamine transients along with the NMDAR antagonism precluded the induction of synaptic plasticity in the VTA and the nucleus accumbens, and did not elicit locomotor sensitization or uncontrolled self-administration. In summary, the dual action of ketamine leads to a unique constellation of dopamine-driven positive reinforcement, but low addiction liability.


Subject(s)
Ketamine , Substance-Related Disorders , Animals , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Ketamine/adverse effects , Ketamine/pharmacology , Mice , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Reinforcement, Psychology , Self Administration , Substance-Related Disorders/etiology , Substance-Related Disorders/prevention & control , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects
20.
Development ; 151(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39162246

ABSTRACT

Marcella Birtele completed her PhD in Malin Parmar's lab at Lund University, Sweden, where her project involved characterising stem cell-derived dopaminergic neurons with the long-term goal of transplanting these neurons into patients with Parkinson's disease. She is now a postdoc at the University of Southern California, USA, where she has been using cortical organoids to explore the role of autism spectrum disorder-associated genes in neural development. Marcella was selected as one of our 2024 PI fellows, a group of researchers who will be supported by Development's Pathway to Independence Programme as they aim to secure independent positions. We spoke to Marcella to hear why she decided to apply to the programme, and how the research questions she hopes to address with her own lab will bridge her previous neurodegeneration and neural development studies.


Subject(s)
Dopaminergic Neurons , Humans , Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , History, 21st Century , History, 20th Century , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Animals , Organoids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL