Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
J Neurosci ; 2021 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-34074735

RESUMO

Repeated cocaine exposure causes dendritic spine loss in the orbitofrontal cortex, which might contribute to poor orbitofrontal cortical function following drug exposure. One challenge, however, has been verifying links between neuronal structural plasticity and behavior, if any. Here we report that cocaine self-administration triggers the loss of dendritic spines on excitatory neurons in the orbitofrontal cortex of male and female mice (as has been reported in rats). To understand functional consequences, we locally ablated neuronal ß1-integrins, cell adhesion receptors that adhere cells to the extracellular matrix and thus support dendritic spine stability. Degradation of ß1-integrin tone: 1) caused dendritic spine loss; 2) exaggerated cocaine-seeking responses in a cue-induced reinstatement test; and 3) impaired the ability of mice to integrate new learning into familiar routines - a key function of the orbitofrontal cortex. Stimulating Abl-related gene (Arg) kinase, over-expressing Proline-rich tyrosine kinase (Pyk2), and inhibiting Rho-associated coiled-coil containing kinase (ROCK) corrected response strategies, uncovering a ß1-integrin-mediated signaling axis that controls orbitofrontal cortical function. Finally, use of a combinatorial gene silencing/chemogenetic strategy revealed that ß1-integrins support the ability of mice to integrate new information into established behaviors by sustaining orbitofrontal cortical connections with the basolateral amygdala.SIGNIFICANCE STATEMENTCocaine degenerates dendritic spines in the orbitofrontal cortex, a region of the brain involved in interlacing new information into established behaviors. One challenge has been verifying links between cellular structural stability and behavior, if any. In this second of two related investigations, we study integrin family receptors, which adhere cells to the extracellular matrix and thereby stabilize dendritic spines (see also DePoy et al., 2019, Journal of Neuroscience). We reveal that ß1-integrins in the orbitofrontal cortex control food- and cocaine-seeking behaviors. For instance, ß1-integrin loss amplifies cocaine-seeking behavior and impairs the ability of mice to integrate new learning into familiar routines. We identify likely intracellular signaling partners by which ß1-integrins support orbitofrontal cortical function and connectivity with the basolateral amygdala.

2.
Dev Neurosci ; 43(6): 376-382, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34695821

RESUMO

Calcium-dependent activator protein for secretion 2 (CAPS2; also referred to as CADPS2) is a dense core vesicle-associated protein that promotes the activity-dependent release of neuropeptides including neurotrophins. Addictive drugs appear to prime neurotrophin release in multiple brain regions, but mechanistic factors are still being elucidated. Here, experimenters administered cocaine to adolescent mice at doses that potentiated later cocaine self-administration. Experimenter-administered cocaine elevated the CAPS2 protein content in the orbitofrontal cortex (OFC; but not striatum) multiple weeks after drug exposure. Meanwhile, proteins that are sensitive to brain-derived neurotrophic factor (BDNF) release and binding (phosphorylated protein kinase B and phosphoinositide 3-kinase, and GABAAα1 levels) did not differ between cocaine-exposed and naive mice in the OFC. This pattern is consistent with evidence that CAPS2 primes stimulated release of neurotrophins like BDNF, rather than basal levels. Thus, cocaine administered at behaviorally relevant doses elevates CAPS2 protein content in the OFC, and the effects are detected long after cocaine exposure.


Assuntos
Cocaína , Proteínas do Tecido Nervoso , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Calmodulina , Cocaína/farmacologia , Vesículas de Núcleo Denso , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Fosfatidilinositol 3-Quinases , Córtex Pré-Frontal/metabolismo
3.
J Neurosci Res ; 98(6): 1020-1030, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31820488

RESUMO

Goal-directed action refers to selecting behaviors based on the expectation that they will be reinforced with desirable outcomes. It is typically conceptualized as opposing habit-based behaviors, which are instead supported by stimulus-response associations and insensitive to consequences. The prelimbic prefrontal cortex (PL) is positioned along the medial wall of the rodent prefrontal cortex. It is indispensable for action-outcome-driven (goal-directed) behavior, consolidating action-outcome relationships and linking contextual information with instrumental behavior. In this brief review, we will discuss the growing list of molecular factors involved in PL function. Ventral to the PL is the medial orbitofrontal cortex (mOFC). We will also summarize emerging evidence from rodents (complementing existing literature describing humans) that it too is involved in action-outcome conditioning. We describe experiments using procedures that quantify responding based on reward value, the likelihood of reinforcement, or effort requirements, touching also on experiments assessing food consumption more generally. We synthesize these findings with the argument that the mOFC is essential to goal-directed action when outcome value information is not immediately observable and must be recalled and inferred.


Assuntos
Comportamento Animal/fisiologia , Objetivos , Córtex Pré-Frontal/fisiologia , Animais , Vias Neurais/fisiologia , Recompensa , Roedores
4.
Cell Rep ; 40(11): 111334, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36103822

RESUMO

In everyday life, we mentally represent possible consequences of our behaviors and integrate specific outcome values into existing knowledge to inform decisions. The medial orbitofrontal cortex (MO) is necessary to adapt behaviors when outcomes are not immediately available-when they and their values need to be envisioned. Nevertheless, neurobiological mechanisms remain unclear. We find that the neuroplasticity-associated neurotrophin receptor tropomyosin receptor kinase B (TrkB) is necessary for mice to integrate outcome-specific value information into choice behavior. This function appears attributable to memory updating (and not retrieval) and the stabilization of dendritic spines on excitatory MO neurons, which led us to investigate inputs to the MO. Ventral hippocampal (vHC)-to-MO projections appear conditionally necessary for value updating, involved in long-term aversion-based value memory updating. Furthermore, vHC-MO-mediated control of choice is TrkB dependent. Altogether, we reveal a vHC-MO connection by which specific value memories are updated, and we position TrkB within this functional circuit.


Assuntos
Hipocampo , Fatores de Crescimento Neural , Animais , Hipocampo/fisiologia , Camundongos , Plasticidade Neuronal/fisiologia , Córtex Pré-Frontal/fisiologia , Transdução de Sinais
5.
Cells ; 10(12)2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34943950

RESUMO

In day-to-day life, we often choose between pursuing familiar behaviors that have been rewarded in the past or adjusting behaviors when new strategies might be more fruitful. The dorsomedial striatum (DMS) is indispensable for flexibly arbitrating between old and new behavioral strategies. The way in which DMS neurons host stable connections necessary for sustained flexibility is still being defined. An entry point to addressing this question may be the structural scaffolds on DMS neurons that house synaptic connections. We find that the non-receptor tyrosine kinase Proline-rich tyrosine kinase 2 (Pyk2) stabilizes both dendrites and spines on striatal medium spiny neurons, such that Pyk2 loss causes dendrite arbor and spine loss. Viral-mediated Pyk2 silencing in the DMS obstructs the ability of mice to arbitrate between rewarded and non-rewarded behaviors. Meanwhile, the overexpression of Pyk2 or the closely related focal adhesion kinase (FAK) enhances this ability. Finally, experiments using combinatorial viral vector strategies suggest that flexible, Pyk2-dependent action involves inputs from the medial prefrontal cortex (mPFC), but not the ventrolateral orbitofrontal cortex (OFC). Thus, Pyk2 stabilizes the striatal medium spiny neuron structure, likely providing substrates for inputs, and supports the capacity of mice to arbitrate between novel and familiar behaviors, including via interactions with the medial-prefrontal cortex.


Assuntos
Quinase 1 de Adesão Focal/genética , Quinase 2 de Adesão Focal/genética , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Animais , Corpo Estriado/metabolismo , Dendritos/genética , Dendritos/metabolismo , Espinhas Dendríticas/genética , Espinhas Dendríticas/metabolismo , Humanos , Camundongos , Neostriado/metabolismo , Neurônios/patologia , Transmissão Sináptica/genética
6.
Behav Brain Res ; 381: 112414, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-31891742

RESUMO

Resistant and generalized fear are hallmark symptoms of Post-Traumatic Stress Disorder (PTSD). Given PTSD is highly comorbid with addiction disorders indicates a maladaptive interaction between fear and reward circuits. To investigate learning processes underlying fear, reward and safety, we trained male rats to discriminate among a fear cue paired with footshock, a reward cue paired with sucrose and an explicit safety cue co-occurring with the fear cue in which no footshocks were delivered. In an attempt to emulate aspects of PTSD, we pre-exposed male rats to a stressor (15 unsignaled footshocks) before training them to fear, reward and safety cues, and subsequent fear and reward extinction. Prior stress did not produce any significant impairments on conditioned inhibition to a safety cue compared to non-stressed controls. However, in subsequent fear extinction, prior stress profoundly impaired fear reduction to an extinguished fear cue. Prior stress also significantly reduced reward seeking to a reward-associated cue throughout training. Together, our data show that prior stress did not affect conditioned inhibition of fear to the same extent as impairing fear extinction. These results have interesting implications on how safety circuits are organized and impacted by stress, leading to possibly new avenues of research on mechanisms of stress disorders, such as PTSD.


Assuntos
Condicionamento Psicológico , Extinção Psicológica , Medo , Inibição Psicológica , Estresse Psicológico/fisiopatologia , Animais , Modelos Animais de Doenças , Ratos , Ratos Long-Evans , Recompensa , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Transtornos de Estresse Pós-Traumáticos/psicologia
7.
Neurotherapeutics ; 17(1): 165-177, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31218603

RESUMO

Cocaine use during adolescence decreases the likelihood that individuals will seek treatment for recurrent drug use. In rodents, developmental cocaine exposure weakens action-consequence decision-making, causing a deferral to familiar, habit-like behavioral response strategies. Here, we aimed to improve action-outcome decision-making. We found that acute pharmacological stimulation of the tyrosine/tropomyosin receptor kinase B (trkB) via 7,8-dihydroxyflavone (7,8-DHF) or 3,4-methylenedioxymethamphetamine (MDMA) blocked cocaine-induced habit biases by strengthening memory for action-outcome associations. We believe that MDMA acts by stimulating neurotrophin/trkB systems in the orbitofrontal cortex (OFC), a region involved in prospectively evaluating the consequences of one's action, because 1) MDMA also increased brain-derived neurotrophic factor (BDNF) in the OFC, 2) MDMA corrected habit biases due to Bdnf loss in the OFC, and 3) overexpression of a truncated isoform of trkB occluded the memory-enhancing effects of MDMA. Thus, selecting actions based on their consequences requires BDNF-trkB in the OFC, the stimulation of which may improve goal attainment in both drug-naïve and cocaine-exposed individuals. SIGNIFICANCE STATEMENT: Cocaine use during adolescence decreases the likelihood that individuals will seek treatment for recurrent drug use, even as adults. Understanding how early-life cocaine exposure impacts goal-oriented action and prospective decision-making in adulthood is thus important. One key aspect of goal-directed decision-making is anticipating the consequences of one's actions, a process that likely involves the orbitofrontal cortex (OFC). In rodents, developmental cocaine exposure weakens action-consequence decision-making, causing a deferral to familiar, habit-like behavioral response strategies. Here, we report that we can improve memory for action-consequence relationships by stimulating neurotrophic factors, which support cell survival, development, and plasticity in the brain. With strengthened action-consequence associations, cocaine-exposed mice regain the ability to optimally select actions based on their likely outcomes. Brain region-selective manipulations reveal that neurotrophin systems in the OFC are necessary for stable memory of action-consequence relationships.


Assuntos
Cocaína/administração & dosagem , Tomada de Decisões/efeitos dos fármacos , Tomada de Decisões/fisiologia , Fatores de Crescimento Neural/fisiologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Feminino , Flavonas/administração & dosagem , Hábitos , Masculino , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/fisiologia , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos Endogâmicos C57BL , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , Proteínas Tirosina Quinases/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA