Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biol Psychiatry ; 93(6): 512-523, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36494220

RESUMEN

BACKGROUND: Obesity is a chronic relapsing disorder that is caused by an excess of caloric intake relative to energy expenditure. There is growing recognition that food motivation is altered in people with obesity. However, it remains unclear how brain circuits that control food motivation are altered in obese animals. METHODS: Using a novel behavioral assay that quantifies work during food seeking, in vivo and ex vivo cell-specific recordings, and a synaptic blocking technique, we tested the hypothesis that activity of circuits promoting appetitive behavior in the core of the nucleus accumbens (NAc) is enhanced in the obese state, particularly during food seeking. RESULTS: We first confirmed that mice made obese with ad libitum exposure to a high fat diet work harder than lean mice to obtain food, consistent with an increase in food motivation in obese mice. We observed greater activation of D1 receptor-expressing NAc spiny projection neurons (NAc D1SPNs) during food seeking in obese mice relative to lean mice. This enhanced activity was not observed in D2 receptor-expressing neurons (D2SPNs). Consistent with these in vivo findings, both intrinsic excitability and excitatory drive onto D1SPNs were enhanced in obese mice relative to lean mice, and these measures were selective for D1SPNs. Finally, blocking synaptic transmission from D1SPNs, but not D2SPNs, in the NAc core decreased physical work during food seeking and, critically, attenuated high fat diet-induced weight gain. CONCLUSIONS: These experiments demonstrate the necessity of NAc core D1SPNs in food motivation and the development of diet-induced obesity, establishing these neurons as a potential therapeutic target for preventing obesity.


Asunto(s)
Motivación , Núcleo Accumbens , Ratones , Animales , Núcleo Accumbens/fisiología , Ratones Obesos , Neuronas/fisiología , Obesidad , Receptores de Dopamina D1/metabolismo , Ratones Endogámicos C57BL
2.
Nat Neurosci ; 25(9): 1124-1128, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36042311

RESUMEN

Fiber photometry enables recording of population neuronal calcium dynamics in awake mice. While the popularity of fiber photometry has grown in recent years, it remains unclear whether photometry reflects changes in action potential firing (that is, 'spiking') or other changes in neuronal calcium. In microscope-based calcium imaging, optical and analytical approaches can help differentiate somatic from neuropil calcium. However, these approaches cannot be readily applied to fiber photometry. As such, it remains unclear whether the fiber photometry signal reflects changes in somatic calcium, changes in nonsomatic calcium or a combination of the two. Here, using simultaneous in vivo extracellular electrophysiology and fiber photometry, along with in vivo endoscopic one-photon and two-photon calcium imaging, we determined that the striatal fiber photometry does not reflect spiking-related changes in calcium and instead primarily reflects nonsomatic changes in calcium.


Asunto(s)
Calcio , Cuerpo Estriado , Potenciales de Acción , Animales , Cuerpo Estriado/fisiología , Ratones , Neuronas/fisiología , Fotometría/métodos
3.
Int Rev Neurobiol ; 159: 187-219, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34446246

RESUMEN

Optogenetic tools allow for the selective activation, inhibition or modulation of genetically-defined neural circuits with incredible temporal precision. Over the past decade, application of these tools in preclinical models of psychiatric disease has advanced our understanding the neural circuit basis of maladaptive behaviors in these disorders. Despite their power as an investigational tool, optogenetics cannot yet be applied in the clinical for the treatment of neurological and psychiatric disorders. To date, deep brain stimulation (DBS) is the only clinical treatment that can be used to achieve circuit-specific neuromodulation in the context of psychiatric. Despite its increasing clinical indications, the mechanisms underlying the therapeutic effects of DBS for psychiatric disorders are poorly understood, which makes optimization difficult. We discuss the variety of optogenetic tools available for preclinical research, and how these tools have been leveraged to reverse-engineer the mechanisms underlying DBS for movement and compulsive disorders. We review studies that have used optogenetics to induce plasticity within defined basal ganglia circuits, to alter neural circuit function and evaluate the corresponding effects on motor and compulsive behaviors. While not immediately applicable to patient populations, the translational power of optogenetics is in inspiring novel DBS protocols by providing a rationale for targeting defined neural circuits to ameliorate specific behavioral symptoms, and by establishing optimal stimulation paradigms that could selectively compensate for pathological synaptic plasticity within these defined neural circuits.


Asunto(s)
Trastornos Mentales , Plasticidad Neuronal , Optogenética , Ganglios Basales/fisiología , Humanos , Trastornos Mentales/fisiopatología , Trastornos Mentales/terapia , Plasticidad Neuronal/fisiología
4.
Elife ; 102021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33779547

RESUMEN

Feeding is critical for survival, and disruption in the mechanisms that govern food intake underlies disorders such as obesity and anorexia nervosa. It is important to understand both food intake and food motivation to reveal mechanisms underlying feeding disorders. Operant behavioral testing can be used to measure the motivational component to feeding, but most food intake monitoring systems do not measure operant behavior. Here, we present a new solution for monitoring both food intake and motivation in rodent home-cages: the Feeding Experimentation Device version 3 (FED3). FED3 measures food intake and operant behavior in rodent home-cages, enabling longitudinal studies of feeding behavior with minimal experimenter intervention. It has a programmable output for synchronizing behavior with optogenetic stimulation or neural recordings. Finally, FED3 design files are open-source and freely available, allowing researchers to modify FED3 to suit their needs.


Obesity and anorexia nervosa are two health conditions related to food intake. Researchers studying these disorders in animal models need to both measure food intake and assess behavioural factors: that is, why animals seek and consume food. Measuring an animal's food intake is usually done by weighing food containers. However, this can be inaccurate due to the small amount of food that rodents eat. As for studying feeding motivation, this can involve calculating the number of times an animal presses a lever to receive a food pellet. These tests are typically conducted in hour-long sessions in temporary testing cages, called operant boxes. Yet, these tests only measure a brief period of a rodent's life. In addition, it takes rodents time to adjust to these foreign environments, which can introduce stress and may alter their feeding behaviour. To address this, Matikainen-Ankney, Earnest, Ali et al. developed a device for monitoring food intake and feeding behaviours around the clock in rodent home cages with minimal experimenter intervention. This 'Feeding Experimentation Device' (FED3) features a pellet dispenser and two 'nose-poke' sensors to measure total food intake, as well as motivation for and learning about food rewards. The battery-powered, wire-free device fits in standard home cages, enabling long-term studies of feeding behaviour with minimal intervention from investigators and less stress on the animals. This means researchers can relate data to circadian rhythms and meal patterns, as Matikainen-Ankney did here. Moreover, the device software is open-source so researchers can customise it to suit their experimental needs. It can also be programmed to synchronise with other instruments used in animal experiments, or across labs running the same behavioural tasks for multi-site studies. Used in this way, it could help improve reproducibility and reliability of results from such studies. In summary, Matikainen-Ankney et al. have presented a new practical solution for studying food-related behaviours in mice and rats. Not only could the device be useful to researchers, it may also be suitable to use in educational settings such as teaching labs and classrooms.


Asunto(s)
Crianza de Animales Domésticos , Condicionamiento Operante , Diseño de Equipo/instrumentación , Conducta Alimentaria , Vivienda para Animales , Roedores/fisiología , Animales , Ingestión de Alimentos , Femenino , Masculino , Ratones
5.
Nat Neurosci ; 24(3): 379-390, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33495635

RESUMEN

The nucleus accumbens shell (NAcSh) and the ventral pallidum (VP) are critical for reward processing, although the question of how coordinated activity within these nuclei orchestrates reward valuation and consumption remains unclear. Inhibition of NAcSh firing is necessary for reward consumption, but the source of this inhibition remains unknown. Here, we report that a subpopulation of VP neurons, the ventral arkypallidal (vArky) neurons, project back to the NAcSh, where they inhibit NAcSh neurons in vivo in mice. Consistent with this pathway driving reward consumption via inhibition of the NAcSh, calcium activity of vArky neurons scaled with reward palatability (which was dissociable from reward seeking) and predicted the subsequent drinking behavior during a free-access paradigm. Activation of the VP-NAcSh pathway increased ongoing reward consumption while amplifying hedonic reactions to reward. These results establish a pivotal role for vArky neurons in the promotion of reward consumption through modulation of NAcSh firing in a value-dependent manner.


Asunto(s)
Potenciales de Acción/fisiología , Prosencéfalo Basal/fisiología , Inhibición Neural/fisiología , Neuronas/fisiología , Recompensa , Animales , Calcio/metabolismo , Conducta de Ingestión de Líquido/fisiología , Femenino , Masculino , Ratones , Vías Nerviosas/fisiología , Núcleo Accumbens/fisiología , Gusto/fisiología
7.
Nat Commun ; 11(1): 6218, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33277492

RESUMEN

Marked deficits in glucose availability, or glucoprivation, elicit organism-wide counter-regulatory responses whose purpose is to restore glucose homeostasis. However, while catecholamine neurons of the ventrolateral medulla (VLMCA) are thought to orchestrate these responses, the circuit and cellular mechanisms underlying specific counter-regulatory responses are largely unknown. Here, we combined anatomical, imaging, optogenetic and behavioral approaches to interrogate the circuit mechanisms by which VLMCA neurons orchestrate glucoprivation-induced food seeking behavior. Using these approaches, we found that VLMCA neurons form functional connections with nucleus accumbens (NAc)-projecting neurons of the posterior portion of the paraventricular nucleus of the thalamus (pPVT). Importantly, optogenetic manipulations revealed that while activation of VLMCA projections to the pPVT was sufficient to elicit robust feeding behavior in well fed mice, inhibition of VLMCA-pPVT communication significantly impaired glucoprivation-induced feeding while leaving other major counterregulatory responses intact. Collectively our findings identify the VLMCA-pPVT-NAc pathway as a previously-neglected node selectively controlling glucoprivation-induced food seeking. Moreover, by identifying the ventrolateral medulla as a direct source of metabolic information to the midline thalamus, our results support a growing body of literature on the role of the PVT in homeostatic regulation.


Asunto(s)
Catecolaminas/metabolismo , Conducta Alimentaria/fisiología , Glucosa/metabolismo , Bulbo Raquídeo/fisiología , Neuronas/fisiología , Núcleos Talámicos Ventrales/fisiología , Animales , Femenino , Homeostasis/fisiología , Masculino , Bulbo Raquídeo/citología , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleos Talámicos de la Línea Media/citología , Núcleos Talámicos de la Línea Media/fisiología , Neuronas/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/fisiología , Núcleos Talámicos Ventrales/citología
8.
Curr Biol ; 30(22): R1366-R1368, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33202234

RESUMEN

The dorsal striatum is important for motor control. Yet whether that control encompasses procedural memories, kinematic refinement, or both is still debated. A recent study has shed new light on the role of the dorsal striatum in learned movement sequences and the effort required to refine them.


Asunto(s)
Cuerpo Estriado , Memoria , Aprendizaje
9.
J Neurophysiol ; 123(6): 2382-2389, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32374202

RESUMEN

Parkinson's disease (PD) risk is increased by stress and certain gene mutations, including the most prevalent PD-linked mutation LRRK2-G2019S. Both PD and stress increase risk for psychiatric symptoms, yet it is unclear how PD-risk genes alter neural circuitry in response to stress that may promote psychopathology. Here we show significant differences between adult G2019S knockin and wild-type (wt) mice in stress-induced behaviors, with an unexpected uncoupling of depression-like and hedonia-like responses in G2019S mice. Moreover, mutant spiny projection neurons in nucleus accumbens (NAc) lack an adaptive, stress-induced change in excitability displayed by wt neurons, and instead show stress-induced changes in synaptic properties that wt neurons lack. Some synaptic alterations in NAc are already evident early in postnatal life. Thus G2019S alters the magnitude and direction of behavioral responses to stress that may reflect unique modifications of adaptive plasticity in cells and circuits implicated in psychopathology in humans.NEW & NOTEWORTHY Depression is associated with Parkinson's disease (PD), and environmental stress is a risk factor for both. We investigated how LRRK2-G2019S PD mutation affects depression-like behaviors, synaptic function, and intrinsic neuronal excitability following stress. In response to stress, the mutation drives abnormal synaptic changes, prevents adaptive changes in intrinsic excitability, and leads to aberrant behaviors, thus defining new ways in which PD mutations derail adaptive plasticity in response to stress that may contribute to disease onset.


Asunto(s)
Conducta Animal , Depresión , Fenómenos Electrofisiológicos , Potenciales Postsinápticos Excitadores , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Núcleo Accumbens , Enfermedad de Parkinson , Estrés Psicológico , Animales , Conducta Animal/fisiología , Depresión/etiología , Depresión/genética , Depresión/fisiopatología , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleo Accumbens/fisiopatología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Estrés Psicológico/complicaciones , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología
10.
Nat Neurosci ; 23(5): 638-650, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284606

RESUMEN

Heightened aggression is characteristic of multiple neuropsychiatric disorders and can have various negative effects on patients, their families and the public. Recent studies in humans and animals have implicated brain reward circuits in aggression and suggest that, in subsets of aggressive individuals, domination of subordinate social targets is reinforcing. In this study, we showed that, in male mice, orexin neurons in the lateral hypothalamus activated a small population of glutamic acid decarboxylase 2 (GAD2)-expressing neurons in the lateral habenula (LHb) via orexin receptor 2 (OxR2) and that activation of these GAD2 neurons promoted male-male aggression and conditioned place preference for aggression-paired contexts. Moreover, LHb GAD2 neurons were inhibitory within the LHb and dampened the activity of the LHb as a whole. These results suggest that the orexin system is important for the regulation of inter-male aggressive behavior and provide the first functional evidence of a local inhibitory circuit within the LHb.


Asunto(s)
Agresión/fisiología , Neuronas GABAérgicas/metabolismo , Habénula/metabolismo , Orexinas/metabolismo , Animales , Masculino , Ratones , Transducción de Señal/fisiología
11.
Obesity (Silver Spring) ; 28(5): 851-856, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32133782

RESUMEN

OBJECTIVE: While changes in diet often result in short-term weight loss, weight loss is not typically maintained. It remains unclear why long-term weight loss is so difficult. It was hypothesized that obesity produces persistent changes in behavior that bias animals toward weight regain after weight loss. METHODS: Mice were induced to gain weight with a high-fat diet for 6 weeks and then induced to lose this weight with a low-fat diet for 7 subsequent weeks. A control group was maintained on the low-fat diet for all 13 weeks. Activity was measured continuously with home cage activity monitors for the entire experiment. Motivation for sweetened food pellets was tested following weight loss. A separate group of mice was reexposed to a high-fat diet following 2, 4, or 8 weeks of withdrawal to assess the rate of weight regain. RESULTS: Activity levels decreased as animals gained weight and partially recovered following weight loss. Motivation for sucrose pellets was persistently heightened after weight loss. Consistent with these behavioral changes, mice also regained weight at a faster rate when reexposed to a high-fat diet after a period of weight loss. CONCLUSIONS: Weight loss after obesity was associated with increased motivation for palatable food and an increased rate of weight regain.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Obesidad/dietoterapia , Aumento de Peso/fisiología , Pérdida de Peso/fisiología , Animales , Femenino , Masculino , Ratones , Motivación
12.
J Neurosci ; 40(8): 1679-1688, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31953369

RESUMEN

The striatum is critical for controlling motor output. However, it remains unclear how striatal output neurons encode and facilitate movement. A prominent theory suggests that striatal units encode movements in bursts of activity near specific events, such as the start or end of actions. These bursts are theorized to gate or permit specific motor actions, thereby encoding and facilitating complex sequences of actions. An alternative theory has suggested that striatal neurons encode continuous changes in sensory or motor information with graded changes in firing rate. Supporting this theory, many striatal neurons exhibit such graded changes without bursting near specific actions. Here, we evaluated these two theories in the same recordings of mice (both male and female). We recorded single-unit and multiunit activity from the dorsomedial striatum of mice as they spontaneously explored an arena. We observed both types of encoding, although continuous encoding was more prevalent than bursting near movement initiation or termination. The majority of recorded units did not exhibit positive linear relationships with speed but instead exhibited nonlinear relationships that peaked at a range of locomotor speeds. Bulk calcium recordings of identified direct and indirect pathway neurons revealed similar speed tuning profiles, indicating that the heterogeneity in response profiles was not due to this genetic distinction. We conclude that continuous encoding of speed is a central component of movement encoding in the striatum.SIGNIFICANCE STATEMENT The striatum is a structure that is linked to volitional movements and is a primary site of pathology in movement disorders. It remains unclear how striatal neurons encode motor parameters and use them to facilitate movement. Here, we evaluated two models for this: a "discrete encoding model" in which striatal neurons facilitate movements with brief burst of activity near the start and end of movements, and a "continuous encoding model," in which striatal neurons encode the sensory or motor state of the animal with continuous changes in firing. We found evidence primarily in support of the continuous encoding model. This may have implications for understanding the striatal control of movement, as well as informing therapeutic approaches for treating movement disorders.


Asunto(s)
Cuerpo Estriado/fisiología , Conducta Exploratoria/fisiología , Movimiento/fisiología , Neuronas/fisiología , Potenciales de Acción/fisiología , Animales , Femenino , Masculino , Ratones
13.
eNeuro ; 6(4)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31235468

RESUMEN

Physical activity is a critical behavioral variable in many research studies and is, therefore, important to quantify. However, existing methods for measuring physical activity have limitations which include high expense, specialized caging or equipment, and high computational overhead. To address these limitations, we present an open-source, cost-effective, device for measuring rodent activity. Our device is battery powered and designed to be placed in vivarium home cages to enable high-throughput, long-term operation with minimal investigator intervention. The primary aim of this study was to assess the feasibility of using passive infrared (PIR) sensors and microcontroller-based dataloggers in a rodent home cages to collect physical activity records. To this end, we developed an open-source PIR based data-logging device called the rodent activity detector (RAD). We publish the design files and code so others can readily build the RAD in their own labs. To demonstrate its utility, we used the RAD to collect physical activity data from 40 individually housed mice for up to 10 weeks. This dataset demonstrates the ability of the RAD to (1) operate in a high-throughput installation, (2) detect high-fat diet (HFD)-induced changes in physical activity, and (3) quantify circadian rhythms in individual animals. We further validated the data output of the RAD with simultaneous video tracking of mice in multiple caging configurations, to determine the features of physical activity that it detects. The RAD is easy to build, economical, and fits in vivarium caging. The scalability of such devices will enable high-throughput studies of physical activity in research studies.


Asunto(s)
Automatización de Laboratorios/instrumentación , Automatización de Laboratorios/métodos , Conducta Animal , Actividad Motora , Animales , Diseño de Equipo , Rayos Infrarrojos , Masculino , Ratones Endogámicos C57BL , Reconocimiento de Normas Patrones Automatizadas
14.
Biochem Soc Trans ; 46(6): 1697-1705, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30514770

RESUMEN

LRRK2 mutation is the most common inherited, autosomal dominant cause of Parkinson's disease (PD) and has also been observed in sporadic cases. Most mutations result in increased LRRK2 kinase activity. LRRK2 is highly expressed in brain regions that receive dense, convergent innervation by dopaminergic and glutamatergic axons, and its levels rise developmentally coincident with glutamatergic synapse formation. The onset and timing of expression suggests strongly that LRRK2 regulates the development, maturation and function of synapses. Several lines of data in mice show that LRRK2-G2019S, the most common LRRK2 mutation, produces an abnormal gain of pathological function that affects synaptic activity, spine morphology, persistent forms of synapse plasticity and behavioral responses to social stress. Effects of the mutation can be detected as early as the second week of postnatal development and can last or have consequences that extend into adulthood and occur in the absence of dopamine loss. These data suggest that the generation of neural circuits that support complex behaviors is modified by LRRK2-G2019S. Whether such alterations impart vulnerability to neurons directly or indirectly, they bring to the forefront the idea that neural circuits within which dopamine neurons eventually degenerate are assembled and utilized in ways that are distinct from circuits that lack this mutation and may contribute to non-motor symptoms observed in humans with PD.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Enfermedad de Parkinson/metabolismo , Animales , Humanos , Mutación/genética
15.
J Neurosci ; 38(45): 9700-9711, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30249796

RESUMEN

The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is a prevalent cause of late-onset Parkinson's disease, producing psychiatric and motor symptoms, including depression, that are indistinguishable from sporadic cases. Here we tested how this mutation impacts depression-related behaviors and associated synaptic responses and plasticity in mice expressing a Lrrk2-G2019S knock-in mutation. Young adult male G2019S knock-in and wild-type mice were subjected to chronic social defeat stress (CSDS), a validated depression model, and other tests of anhedonia, anxiety, and motor learning. We found that G2019S mice were highly resilient to CSDS, failing to exhibit social avoidance compared to wild-type mice, many of which exhibited prominent social avoidance and were thus susceptible to CSDS. In the absence of CSDS, no behavioral differences between genotypes were found. Whole-cell recordings of spiny projection neurons (SPNs) in the nucleus accumbens revealed that glutamatergic synapses in G2019S mice lacked functional calcium-permeable AMPARs, and following CSDS, failed to accumulate inwardly rectifying AMPAR responses characteristic of susceptible mice. Based on this abnormal AMPAR response profile, we asked whether long-term potentiation (LTP) of corticostriatal synaptic strength was affected. We found that both D1 receptor (D1R)- and D2R-SPNs in G2019S mutants were unable to express LTP, with D2R-SPNs abnormally expressing long-term depression following an LTP-induction protocol. Thus, G2019S promotes resilience to chronic social stress in young adulthood, likely reflecting synapses constrained in their ability to undergo experience-dependent plasticity. These unexpected findings may indicate early adaptive coping mechanisms imparted by the G2019S mutation.SIGNIFICANCE STATEMENT The G2019S mutation in LRRK2 causes late-onset Parkinson's disease (PD). LRRK2 is highly expressed in striatal neurons throughout life, but it is unclear how mutant LRRK2 affects striatal neuron function and behaviors in young adulthood. We addressed this question using Lrrk2-G2019S knock-in mice. The data show that young adult G2019S mice were unusually resilient to a depression-like syndrome resulting from chronic social stress. Further, mutant striatal synapses were incapable of forms of synaptic plasticity normally accompanying depression-like behavior and important for supporting the full range of cognitive function. These data suggest that in humans, LRRK2 mutation may affect striatal circuit function in ways that alter normal responses to stress and could be relevant for treatment strategies for non-motor PD symptoms.


Asunto(s)
Relaciones Interpersonales , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Mutación/genética , Plasticidad Neuronal/fisiología , Enfermedad de Parkinson/genética , Resiliencia Psicológica , Estrés Psicológico/genética , Factores de Edad , Animales , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/psicología , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología
16.
Ann N Y Acad Sci ; 1428(1): 221-239, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29741270

RESUMEN

The obesity epidemic is a leading cause of health problems in the United States, increasing the risk of cardiovascular, endocrine, and psychiatric diseases. Although many people lose weight through changes in diet and lifestyle, keeping the weight off remains a challenge. Here, we discuss a hypothesis that seeks to explain why obesity is so persistent. There is a great degree of overlap in the circuits implicated in substance use disorder and obesity, and neural plasticity of these circuits in response to drugs of abuse is well documented. We hypothesize that obesity is also associated with neural plasticity in these circuits, and this may underlie persistent changes in behavior, energy balance, and body weight. Here, we discuss how obesity-associated reductions in motivation and physical activity may be rooted in neurophysiological alterations in these circuits. Such plasticity may alter how humans and animals use, expend, and store energy, even after weight loss.


Asunto(s)
Conducta Alimentaria/fisiología , Plasticidad Neuronal , Obesidad/fisiopatología , Analgésicos Opioides/farmacología , Animales , Comorbilidad , Cuerpo Estriado/fisiopatología , Dieta Alta en Grasa/efectos adversos , Dopamina/fisiología , Metabolismo Energético/fisiología , Ejercicio Físico/fisiología , Ejercicio Físico/psicología , Glutamatos/fisiología , Humanos , Hipotálamo/fisiopatología , Inflamación , Motivación/fisiología , Red Nerviosa/fisiología , Vías Nerviosas/fisiología , Obesidad/epidemiología , Obesidad/metabolismo , Obesidad/psicología , Ratas , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/fisiología , Receptores Opioides/fisiología , Recurrencia , Trastornos Relacionados con Sustancias/epidemiología , Trastornos Relacionados con Sustancias/fisiopatología , Trastornos Relacionados con Sustancias/psicología , Área Tegmental Ventral/fisiopatología , Pérdida de Peso/fisiología
17.
Biol Psychiatry ; 83(12): 1012-1023, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29452828

RESUMEN

BACKGROUND: The ability to appropriately integrate and respond to rewarding and aversive stimuli is essential for survival. The ventral pallidum (VP) plays a critical role in processing both rewarding and aversive stimuli. However, the VP is a heterogeneous structure, and how VP subpopulations integrate into larger reward networks to ultimately modulate these behaviors is not known. We identify a noncanonical population of glutamatergic VP neurons that play a unique role in responding to aversive stimuli and constraining inappropriate reward seeking. METHODS: Using neurochemical, genetic, and electrophysiological approaches, we characterized glutamatergic VP neurons (n = 4-8 mice/group). We performed patch clamp and in vivo electrophysiology recordings in the lateral habenula, rostromedial tegmental nucleus, and ventral tegmental area to determine the effect of glutamatergic VP neuron activation in these target regions (n = 6-10 mice/group). Finally, we selectively optogenetically stimulated glutamatergic VP neurons in a real-time place preference task and ablated these neurons using a virally expressed caspase to determine their necessity for reward seeking. RESULTS: Glutamatergic VP neurons exhibit little overlap with cholinergic or gamma-aminobutyric acidergic markers, the canonical VP subtypes, and exhibit distinct membrane properties. Glutamatergic VP neurons innervate and increase firing activity of the lateral habenula, rostromedial tegmental nucleus, and gamma-aminobutyric acidergic ventral tegmental area neurons. While nonselective optogenetic stimulation of the VP induced a robust place preference, selective activation of glutamatergic VP neurons induced a place avoidance. Viral ablation of glutamatergic VP neurons increased reward responding and abolished taste aversion to sucrose. CONCLUSIONS: Glutamatergic VP neurons constitute a noncanonical subpopulation of VP neurons. These glutamatergic VP neurons increase activity of the lateral habenula, rostromedial tegmental nucleus, and gamma-aminobutyric acidergic ventral tegmental area neurons and adaptively constrain reward seeking.


Asunto(s)
Prosencéfalo Basal/citología , Ácido Glutámico/metabolismo , Habénula/fisiología , Neuronas/fisiología , Recompensa , Área Tegmental Ventral/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Reacción de Prevención/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Condicionamiento Operante/fisiología , Dopamina/metabolismo , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Femenino , Ácido Glutámico/farmacología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Optogenética , Parvalbúminas/genética , Parvalbúminas/metabolismo , Técnicas de Placa-Clamp , Gusto , Transducción Genética , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Proteína Fluorescente Roja
18.
J Neurosci ; 36(27): 7128-41, 2016 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-27383589

RESUMEN

UNLABELLED: Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) can cause Parkinson's disease (PD), and the most common disease-associated mutation, G2019S, increases kinase activity. Because LRRK2 expression levels rise during synaptogenesis and are highest in dorsal striatal spiny projection neurons (SPNs), we tested the hypothesis that the LRRK2-G2019S mutation would alter development of excitatory synaptic networks in dorsal striatum. To circumvent experimental confounds associated with LRRK2 overexpression, we used mice expressing LRRK2-G2019S or D2017A (kinase-dead) knockin mutations. In whole-cell recordings, G2019S SPNs exhibited a fourfold increase in sEPSC frequency compared with wild-type SPNs in postnatal day 21 mice. Such heightened neural activity was increased similarly in direct- and indirect-pathway SPNs, and action potential-dependent activity was particularly elevated. Excitatory synaptic activity in D2017A SPNs was similar to wild type, indicating a selective effect of G2019S. Acute exposure to LRRK2 kinase inhibitors normalized activity, supporting that excessive neural activity in G2019S SPNs is mediated directly and is kinase dependent. Although dendritic arborization and densities of excitatory presynaptic terminals and postsynaptic dendritic spines in G2019S SPNs were similar to wild type, G2019S SPNs displayed larger spines that were matched functionally by a shift toward larger postsynaptic response amplitudes. Acutely isolating striatum from overlying neocortex normalized sEPSC frequency in G2019S mutants, supporting that abnormal corticostriatal activity is involved. These findings indicate that the G2019S mutation imparts a gain-of-abnormal function to SPN activity and morphology during a stage of development when activity can permanently modify circuit structure and function. SIGNIFICANCE STATEMENT: Mutations in the kinase domain of leucine-rich repeat kinase 2 (LRRK2) follow Parkinson's disease (PD) heritability. How such mutations affect brain function is poorly understood. LRRK2 expression levels rise after birth at a time when synapses are forming and are highest in dorsal striatum, suggesting that LRRK2 regulates development of striatal circuits. During a period of postnatal development when activity plays a large role in permanently shaping neural circuits, our data show how the most common PD-causing LRRK2 mutation dramatically alters excitatory synaptic activity and the shape of postsynaptic structures in striatum. These findings provide new insight into early functional and structural aberrations in striatal connectivity that may predispose striatal circuitry to both motor and nonmotor dysfunction later in life.


Asunto(s)
Cuerpo Estriado/patología , Regulación del Desarrollo de la Expresión Génica/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Mutación/genética , Neuronas/fisiología , Enfermedad de Parkinson/genética , Animales , Animales Recién Nacidos , Cuerpo Estriado/fisiopatología , Dendritas/patología , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Femenino , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/patología , Técnicas de Placa-Clamp , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
19.
Hippocampus ; 24(8): 943-962, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24753442

RESUMEN

N-Cadherin and ß-catenin form a transsynaptic adhesion complex required for spine and synapse development. In adulthood, N-cadherin mediates persistent synaptic plasticity, but whether the role of N-cadherin at mature synapses is similar to that at developing synapses is unclear. To address this, we conditionally ablated N-cadherin from excitatory forebrain synapses in mice starting in late postnatal life and examined hippocampal structure and function in adulthood. In the absence of N-cadherin, ß-catenin levels were reduced, but numbers of excitatory synapses were unchanged, and there was no impact on number or shape of dendrites or spines. However, the composition of synaptic molecules was altered. Levels of GluA1 and its scaffolding protein PSD95 were diminished and the density of immunolabeled puncta was decreased, without effects on other glutamate receptors and their scaffolding proteins. Additionally, loss of N-cadherin at excitatory synapses triggered increases in the density of markers for inhibitory synapses and decreased severity of hippocampal seizures. Finally, adult mutant mice were profoundly impaired in hippocampal-dependent memory for spatial episodes. These results demonstrate a novel function for the N-cadherin/ß-catenin complex in regulating ionotropic receptor composition of excitatory synapses, an appropriate balance of excitatory and inhibitory synaptic proteins and the maintenance of neural circuitry necessary to generate flexible yet persistent cognitive and synaptic function.


Asunto(s)
Cadherinas/deficiencia , Hipocampo/fisiopatología , Inhibición Neural/fisiología , Sinapsis/fisiología , beta Catenina/metabolismo , Animales , Cadherinas/genética , Dendritas/fisiología , Espinas Dendríticas/fisiología , Homólogo 4 de la Proteína Discs Large , Guanilato-Quinasas/metabolismo , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Ácido Kaínico , Masculino , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/fisiología , Prosencéfalo/citología , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/fisiopatología , Receptores AMPA/metabolismo , Convulsiones/fisiopatología , Memoria Espacial/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...