Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 17.675
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(14): 3671-3689.e23, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38866017

RESUMEN

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.


Asunto(s)
Supervivencia Celular , Neuronas Dopaminérgicas , FN-kappa B , Factor de Necrosis Tumoral alfa , Proteína p53 Supresora de Tumor , Neuronas Dopaminérgicas/metabolismo , Animales , Humanos , FN-kappa B/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ratones , Supervivencia Celular/efectos de los fármacos , Transducción de Señal , Enfermedad de Parkinson/metabolismo , Células Madre Pluripotentes/metabolismo , Apoptosis , Modelos Animales de Enfermedad , Sistemas CRISPR-Cas
2.
Cell ; 186(18): 3845-3861.e24, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37591240

RESUMEN

Dopaminergic projections regulate various brain functions and are implicated in many neuropsychiatric disorders. There are two anatomically and functionally distinct dopaminergic projections connecting the midbrain to striatum: nigrostriatal, which controls movement, and mesolimbic, which regulates motivation. However, how these discrete dopaminergic synaptic connections are established is unknown. Through an unbiased search, we identify that two groups of antagonistic TGF-ß family members, bone morphogenetic protein (BMP)6/BMP2 and transforming growth factor (TGF)-ß2, regulate dopaminergic synapse development of nigrostriatal and mesolimbic neurons, respectively. Projection-preferential expression of their receptors contributes to specific synapse development. Downstream, Smad1 and Smad2 are specifically activated and required for dopaminergic synapse development and function in nigrostriatal vs. mesolimbic projections. Remarkably, Smad1 mutant mice show motor defects, whereas Smad2 mutant mice show lack of motivation. These results uncover the molecular logic underlying the proper establishment of functionally segregated dopaminergic synapses and may provide strategies to treat relevant, projection-specific disease symptoms by targeting specific BMPs/TGF-ß and/or Smads.


Asunto(s)
Cuerpo Estriado , Dopamina , Animales , Ratones , Mesencéfalo , Motivación , Movimiento , Sinapsis
3.
Cell ; 186(18): 3862-3881.e28, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37572660

RESUMEN

Male sexual behavior is innate and rewarding. Despite its centrality to reproduction, a molecularly specified neural circuit governing innate male sexual behavior and reward remains to be characterized. We have discovered a developmentally wired neural circuit necessary and sufficient for male mating. This circuit connects chemosensory input to BNSTprTac1 neurons, which innervate POATacr1 neurons that project to centers regulating motor output and reward. Epistasis studies demonstrate that BNSTprTac1 neurons are upstream of POATacr1 neurons, and BNSTprTac1-released substance P following mate recognition potentiates activation of POATacr1 neurons through Tacr1 to initiate mating. Experimental activation of POATacr1 neurons triggers mating, even in sexually satiated males, and it is rewarding, eliciting dopamine release and self-stimulation of these cells. Together, we have uncovered a neural circuit that governs the key aspects of innate male sexual behavior: motor displays, drive, and reward.


Asunto(s)
Vías Nerviosas , Conducta Sexual Animal , Animales , Masculino , Neuronas/fisiología , Recompensa , Conducta Sexual Animal/fisiología , Ratones
4.
Cell ; 184(10): 2733-2749.e16, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33861952

RESUMEN

Significant evidence supports the view that dopamine shapes learning by encoding reward prediction errors. However, it is unknown whether striatal targets receive tailored dopamine dynamics based on regional functional specialization. Here, we report wave-like spatiotemporal activity patterns in dopamine axons and release across the dorsal striatum. These waves switch between activational motifs and organize dopamine transients into localized clusters within functionally related striatal subregions. Notably, wave trajectories were tailored to task demands, propagating from dorsomedial to dorsolateral striatum when rewards are contingent on animal behavior and in the opponent direction when rewards are independent of behavioral responses. We propose a computational architecture in which striatal dopamine waves are sculpted by inference about agency and provide a mechanism to direct credit assignment to specialized striatal subregions. Supporting model predictions, dorsomedial dopamine activity during reward-pursuit signaled the extent of instrumental control and interacted with reward waves to predict future behavioral adjustments.


Asunto(s)
Axones/metabolismo , Conducta Animal , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Recompensa , Animales , Femenino , Masculino , Ratones , Ratones Mutantes
5.
Cell ; 184(26): 6344-6360.e18, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34890577

RESUMEN

The anterior insular cortex (aIC) plays a critical role in cognitive and motivational control of behavior, but the underlying neural mechanism remains elusive. Here, we show that aIC neurons expressing Fezf2 (aICFezf2), which are the pyramidal tract neurons, signal motivational vigor and invigorate need-seeking behavior through projections to the brainstem nucleus tractus solitarii (NTS). aICFezf2 neurons and their postsynaptic NTS neurons acquire anticipatory activity through learning, which encodes the perceived value and the vigor of actions to pursue homeostatic needs. Correspondingly, aIC → NTS circuit activity controls vigor, effort, and striatal dopamine release but only if the action is learned and the outcome is needed. Notably, aICFezf2 neurons do not represent taste or valence. Moreover, aIC → NTS activity neither drives reinforcement nor influences total consumption. These results pinpoint specific functions of aIC → NTS circuit for selectively controlling motivational vigor and suggest that motivation is subserved, in part, by aIC's top-down regulation of dopamine signaling.


Asunto(s)
Tronco Encefálico/fisiología , Corteza Insular/fisiología , Motivación , Vías Nerviosas/fisiología , Animales , Conducta Animal , Dopamina/metabolismo , Femenino , Aprendizaje , Masculino , Ratones Endogámicos C57BL , Neuronas/fisiología , Núcleo Accumbens/metabolismo , Factores de Tiempo
6.
Cell ; 184(4): 943-956.e18, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33571432

RESUMEN

Dopamine receptors, including D1- and D2-like receptors, are important therapeutic targets in a variety of neurological syndromes, as well as cardiovascular and kidney diseases. Here, we present five cryoelectron microscopy (cryo-EM) structures of the dopamine D1 receptor (DRD1) coupled to Gs heterotrimer in complex with three catechol-based agonists, a non-catechol agonist, and a positive allosteric modulator for endogenous dopamine. These structures revealed that a polar interaction network is essential for catecholamine-like agonist recognition, whereas specific motifs in the extended binding pocket were responsible for discriminating D1- from D2-like receptors. Moreover, allosteric binding at a distinct inner surface pocket improved the activity of DRD1 by stabilizing endogenous dopamine interaction at the orthosteric site. DRD1-Gs interface revealed key features that serve as determinants for G protein coupling. Together, our study provides a structural understanding of the ligand recognition, allosteric regulation, and G protein coupling mechanisms of DRD1.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Receptores de Dopamina D1/metabolismo , Transducción de Señal , Regulación Alostérica , Sitio Alostérico , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Catecoles/metabolismo , Microscopía por Crioelectrón , Fenoldopam/química , Fenoldopam/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Subunidades alfa de la Proteína de Unión al GTP Gs/ultraestructura , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Multimerización de Proteína , Receptores de Dopamina D1/química , Receptores de Dopamina D1/ultraestructura , Receptores de Dopamina D2/metabolismo , Homología Estructural de Proteína
7.
Cell ; 184(4): 931-942.e18, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33571431

RESUMEN

The D1- and D2-dopamine receptors (D1R and D2R), which signal through Gs and Gi, respectively, represent the principal stimulatory and inhibitory dopamine receptors in the central nervous system. D1R and D2R also represent the main therapeutic targets for Parkinson's disease, schizophrenia, and many other neuropsychiatric disorders, and insight into their signaling is essential for understanding both therapeutic and side effects of dopaminergic drugs. Here, we report four cryoelectron microscopy (cryo-EM) structures of D1R-Gs and D2R-Gi signaling complexes with selective and non-selective dopamine agonists, including two currently used anti-Parkinson's disease drugs, apomorphine and bromocriptine. These structures, together with mutagenesis studies, reveal the conserved binding mode of dopamine agonists, the unique pocket topology underlying ligand selectivity, the conformational changes in receptor activation, and potential structural determinants for G protein-coupling selectivity. These results provide both a molecular understanding of dopamine signaling and multiple structural templates for drug design targeting the dopaminergic system.


Asunto(s)
Receptores de Dopamina D1/química , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/química , Receptores de Dopamina D2/metabolismo , Transducción de Señal , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/análogos & derivados , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Secuencia de Aminoácidos , Secuencia Conservada , Microscopía por Crioelectrón , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Dopamina D1/ultraestructura , Receptores de Dopamina D2/ultraestructura , Homología Estructural de Proteína
8.
Cell ; 182(4): 960-975.e15, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32763155

RESUMEN

Parental behavior is pervasive throughout the animal kingdom and essential for species survival. However, the relative contribution of the father to offspring care differs markedly across animals, even between related species. The mechanisms that organize and control paternal behavior remain poorly understood. Using Sprague-Dawley rats and C57BL/6 mice, two species at opposite ends of the paternal spectrum, we identified that distinct electrical oscillation patterns in neuroendocrine dopamine neurons link to a chain of low dopamine release, high circulating prolactin, prolactin receptor-dependent activation of medial preoptic area galanin neurons, and paternal care behavior in male mice. In rats, the same parameters exhibit inverse profiles. Optogenetic manipulation of these rhythms in mice dramatically shifted serum prolactin and paternal behavior, whereas injecting prolactin into non-paternal rat sires triggered expression of parental care. These findings identify a frequency-tuned brain-endocrine-brain circuit that can act as a gain control system determining a species' parental strategy.


Asunto(s)
Dopamina/metabolismo , Hipotálamo/fisiología , Neuronas/fisiología , Conducta Paterna/fisiología , Animales , Encéfalo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Optogenética , Técnicas de Placa-Clamp , Prolactina/sangre , Ratas , Ratas Sprague-Dawley , Receptores de Prolactina/deficiencia , Receptores de Prolactina/genética , Receptores de Prolactina/metabolismo
9.
Cell ; 181(3): 702-715.e20, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32315619

RESUMEN

Protein phosphatase 2A (PP2A) enzymes can suppress tumors, but they are often inactivated in human cancers overexpressing inhibitory proteins. Here, we identify a class of small-molecule iHAPs (improved heterocyclic activators of PP2A) that kill leukemia cells by allosterically assembling a specific heterotrimeric PP2A holoenzyme consisting of PPP2R1A (scaffold), PPP2R5E (B56ε, regulatory), and PPP2CA (catalytic) subunits. One compound, iHAP1, activates this complex but does not inhibit dopamine receptor D2, a mediator of neurologic toxicity induced by perphenazine and related neuroleptics. The PP2A complex activated by iHAP1 dephosphorylates the MYBL2 transcription factor on Ser241, causing irreversible arrest of leukemia and other cancer cells in prometaphase. In contrast, SMAPs, a separate class of compounds, activate PP2A holoenzymes containing a different regulatory subunit, do not dephosphorylate MYBL2, and arrest tumor cells in G1 phase. Our findings demonstrate that small molecules can serve as allosteric switches to activate distinct PP2A complexes with unique substrates.


Asunto(s)
Proteína Fosfatasa 2/metabolismo , Apoptosis , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Activadores de Enzimas/metabolismo , Fase G1 , Humanos , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , Fenotiazinas/farmacología , Fosforilación , Proteína Fosfatasa 2/fisiología , Subunidades de Proteína/metabolismo , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
10.
Cell ; 181(6): 1364-1379.e14, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32470395

RESUMEN

Small molecule neurotensin receptor 1 (NTSR1) agonists have been pursued for more than 40 years as potential therapeutics for psychiatric disorders, including drug addiction. Clinical development of NTSR1 agonists has, however, been precluded by their severe side effects. NTSR1, a G protein-coupled receptor (GPCR), signals through the canonical activation of G proteins and engages ß-arrestins to mediate distinct cellular signaling events. Here, we characterize the allosteric NTSR1 modulator SBI-553. This small molecule not only acts as a ß-arrestin-biased agonist but also extends profound ß-arrestin bias to the endogenous ligand by selectively antagonizing G protein signaling. SBI-553 shows efficacy in animal models of psychostimulant abuse, including cocaine self-administration, without the side effects characteristic of balanced NTSR1 agonism. These findings indicate that NTSR1 G protein and ß-arrestin activation produce discrete and separable physiological effects, thus providing a strategy to develop safer GPCR-targeting therapeutics with more directed pharmacological action.


Asunto(s)
Conducta Adictiva/metabolismo , Receptores de Neurotensina/metabolismo , beta-Arrestinas/metabolismo , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Animales , Conducta Adictiva/tratamiento farmacológico , Línea Celular , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Bibliotecas de Moléculas Pequeñas/farmacología
11.
Cell ; 178(1): 60-75.e19, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31230716

RESUMEN

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.


Asunto(s)
Aprendizaje por Asociación/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Cuerpos Pedunculados/fisiología , Receptores de Dopamina D1/metabolismo , Receptores Dopaminérgicos/metabolismo , Animales , Conducta Animal/fisiología , Condicionamiento Clásico/fisiología , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Plasticidad Neuronal , Odorantes , Recompensa , Olfato/fisiología , Potenciales Sinápticos/fisiología , Factores de Tiempo
12.
Cell ; 174(2): 481-496.e19, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-30007419

RESUMEN

Dopamine (DA) is a central monoamine neurotransmitter involved in many physiological and pathological processes. A longstanding yet largely unmet goal is to measure DA changes reliably and specifically with high spatiotemporal precision, particularly in animals executing complex behaviors. Here, we report the development of genetically encoded GPCR-activation-based-DA (GRABDA) sensors that enable these measurements. In response to extracellular DA, GRABDA sensors exhibit large fluorescence increases (ΔF/F0 ∼90%) with subcellular resolution, subsecond kinetics, nanomolar to submicromolar affinities, and excellent molecular specificity. GRABDA sensors can resolve a single-electrical-stimulus-evoked DA release in mouse brain slices and detect endogenous DA release in living flies, fish, and mice. In freely behaving mice, GRABDA sensors readily report optogenetically elicited nigrostriatal DA release and depict dynamic mesoaccumbens DA signaling during Pavlovian conditioning or during sexual behaviors. Thus, GRABDA sensors enable spatiotemporally precise measurements of DA dynamics in a variety of model organisms while exhibiting complex behaviors.


Asunto(s)
Dopamina/análisis , Drosophila/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Conducta Animal , Dopamina/metabolismo , Femenino , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Neuronas/citología , Neuronas/metabolismo , Optogenética/métodos , Receptores Acoplados a Proteínas G/genética , Canales Catiónicos TRPV/genética , Proteínas de Pez Cebra/genética
13.
Cell ; 175(3): 665-678.e23, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30245012

RESUMEN

The gut is now recognized as a major regulator of motivational and emotional states. However, the relevant gut-brain neuronal circuitry remains unknown. We show that optical activation of gut-innervating vagal sensory neurons recapitulates the hallmark effects of stimulating brain reward neurons. Specifically, right, but not left, vagal sensory ganglion activation sustained self-stimulation behavior, conditioned both flavor and place preferences, and induced dopamine release from Substantia nigra. Cell-specific transneuronal tracing revealed asymmetric ascending pathways of vagal origin throughout the CNS. In particular, transneuronal labeling identified the glutamatergic neurons of the dorsolateral parabrachial region as the obligatory relay linking the right vagal sensory ganglion to dopamine cells in Substantia nigra. Consistently, optical activation of parabrachio-nigral projections replicated the rewarding effects of right vagus excitation. Our findings establish the vagal gut-to-brain axis as an integral component of the neuronal reward pathway. They also suggest novel vagal stimulation approaches to affective disorders.


Asunto(s)
Intestinos/fisiología , Recompensa , Sustancia Negra/fisiología , Nervio Vago/fisiología , Vías Aferentes/metabolismo , Vías Aferentes/fisiología , Animales , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Ácido Glutámico/metabolismo , Intestinos/inervación , Masculino , Ratones , Ratones Endogámicos C57BL , Optogenética
14.
Cell ; 175(3): 709-722.e15, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30245010

RESUMEN

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.


Asunto(s)
Extinción Psicológica , Memoria , Animales , Conducta Apetitiva , Calcio/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/fisiología , Drosophila melanogaster , Femenino , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/fisiología , Plasticidad Neuronal
15.
Cell ; 174(6): 1436-1449.e20, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30146163

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Neuronas Dopaminérgicas/metabolismo , Proteínas Nucleares/genética , Sinapsis/metabolismo , Transporte Activo de Núcleo Celular , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Señalización del Calcio , Núcleo Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis , Mutación Missense , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
16.
Cell ; 172(4): 706-718.e15, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29398114

RESUMEN

Dopamine controls essential brain functions through volume transmission. Different from fast synaptic transmission, where neurotransmitter release and receptor activation are tightly coupled by an active zone, dopamine transmission is widespread and may not necessitate these organized release sites. Here, we determine whether striatal dopamine secretion employs specialized machinery for release. Using super resolution microscopy, we identified co-clustering of the active zone scaffolding proteins bassoon, RIM and ELKS in ∼30% of dopamine varicosities. Conditional RIM knockout disrupted this scaffold and, unexpectedly, abolished dopamine release, while ELKS knockout had no effect. Optogenetic experiments revealed that dopamine release was fast and had a high release probability, indicating the presence of protein scaffolds for coupling Ca2+ influx to vesicle fusion. Hence, dopamine secretion is mediated by sparse, mechanistically specialized active zone-like release sites. This architecture supports spatially and temporally precise coding for dopamine and provides molecular machinery for regulation.


Asunto(s)
Axones/metabolismo , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Transmisión Sináptica/fisiología , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Cuerpo Estriado/citología , Dopamina/genética , Técnicas de Silenciamiento del Gen , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al GTP rab
17.
Immunity ; 56(2): 320-335.e9, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36693372

RESUMEN

Neuronal signals have emerged as pivotal regulators of group 2 innate lymphoid cells (ILC2s) that regulate tissue homeostasis and allergic inflammation. The molecular pathways underlying the neuronal regulation of ILC2 responses in lungs remain to be fully elucidated. Here, we found that the abundance of neurotransmitter dopamine was negatively correlated with circulating ILC2 numbers and positively associated with pulmonary function in humans. Dopamine potently suppressed lung ILC2 responses in a DRD1-receptor-dependent manner. Genetic deletion of Drd1 or local ablation of dopaminergic neurons augmented ILC2 responses and allergic lung inflammation. Transcriptome and metabolic analyses revealed that dopamine impaired the mitochondrial oxidative phosphorylation (OXPHOS) pathway in ILC2s. Augmentation of OXPHOS activity with oltipraz antagonized the inhibitory effect of dopamine. Local administration of dopamine alleviated allergen-induced ILC2 responses and airway inflammation. These findings demonstrate that dopamine represents an inhibitory regulator of ILC2 responses in allergic airway inflammation.


Asunto(s)
Inmunidad Innata , Neumonía , Humanos , Dopamina/metabolismo , Linfocitos , Pulmón/metabolismo , Neumonía/metabolismo , Inflamación/metabolismo , Interleucina-33/metabolismo
18.
Immunity ; 56(12): 2773-2789.e8, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37992711

RESUMEN

Although the gut microbiota can influence central nervous system (CNS) autoimmune diseases, the contribution of the intestinal epithelium to CNS autoimmunity is less clear. Here, we showed that intestinal epithelial dopamine D2 receptors (IEC DRD2) promoted sex-specific disease progression in an animal model of multiple sclerosis. Female mice lacking Drd2 selectively in intestinal epithelial cells showed a blunted inflammatory response in the CNS and reduced disease progression. In contrast, overexpression or activation of IEC DRD2 by phenylethylamine administration exacerbated disease severity. This was accompanied by altered lysozyme expression and gut microbiota composition, including reduced abundance of Lactobacillus species. Furthermore, treatment with N2-acetyl-L-lysine, a metabolite derived from Lactobacillus, suppressed microglial activation and neurodegeneration. Taken together, our study indicates that IEC DRD2 hyperactivity impacts gut microbial abundances and increases susceptibility to CNS autoimmune diseases in a female-biased manner, opening up future avenues for sex-specific interventions of CNS autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Esclerosis Múltiple , Masculino , Femenino , Ratones , Animales , Esclerosis Múltiple/metabolismo , Modelos Animales de Enfermedad , Transducción de Señal , Progresión de la Enfermedad , Receptores Dopaminérgicos
19.
Cell ; 169(5): 956-969.e17, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28502772

RESUMEN

Animals exhibit a behavioral response to novel sensory stimuli about which they have no prior knowledge. We have examined the neural and behavioral correlates of novelty and familiarity in the olfactory system of Drosophila. Novel odors elicit strong activity in output neurons (MBONs) of the α'3 compartment of the mushroom body that is rapidly suppressed upon repeated exposure to the same odor. This transition in neural activity upon familiarization requires odor-evoked activity in the dopaminergic neuron innervating this compartment. Moreover, exposure of a fly to novel odors evokes an alerting response that can also be elicited by optogenetic activation of α'3 MBONs. Silencing these MBONs eliminates the alerting behavior. These data suggest that the α'3 compartment plays a causal role in the behavioral response to novel and familiar stimuli as a consequence of dopamine-mediated plasticity at the Kenyon cell-MBONα'3 synapse.


Asunto(s)
Drosophila melanogaster/fisiología , Cuerpos Pedunculados/fisiología , Animales , Neuronas Dopaminérgicas/fisiología , Aprendizaje , Memoria , Cuerpos Pedunculados/citología , Odorantes , Olfato
20.
Annu Rev Neurosci ; 46: 359-380, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37068787

RESUMEN

Striosomes form neurochemically specialized compartments of the striatum embedded in a large matrix made up of modules called matrisomes. Striosome-matrix architecture is multiplexed with the canonical direct-indirect organization of the striatum. Striosomal functions remain to be fully clarified, but key information is emerging. First, striosomes powerfully innervate nigral dopamine-containing neurons and can completely shut down their activity, with a following rebound excitation. Second, striosomes receive limbic and cognition-related corticostriatal afferents and are dynamically modulated in relation to value-based actions. Third, striosomes are spatially interspersed among matrisomes and interneurons and are influenced by local and global neuromodulatory and oscillatory activities. Fourth, striosomes tune engagement and the motivation to perform reinforcement learning, to manifest stereotypical behaviors, and to navigate valence conflicts and valence discriminations. We suggest that, at an algorithmic level, striosomes could serve as distributed scaffolds to provide formats of the striatal computations generated through development and refined through learning. We propose that striosomes affect subjective states. By transforming corticothalamic and other inputs to the functional formats of the striatum, they could implement state transitions in nigro-striato-nigral circuits to affect bodily and cognitive actions according to internal motives whose functions are compromised in neuropsychiatric conditions.


Asunto(s)
Ganglios Basales , Volición , Ganglios Basales/fisiología , Cuerpo Estriado/fisiología , Interneuronas , Refuerzo en Psicología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA