Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Adv Drug Alcohol Res ; 4: 12094, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38524847

RESUMEN

Adolescent alcohol drinking is linked to high rates of adult alcohol problems and alcohol use disorder (AUD). The Neurobiology of Alcohol Drinking in Adulthood (NADIA) consortium adolescent intermittent ethanol (AIE) models adolescent binge drinking, followed by abstinent maturation to adulthood to determine the persistent AIE changes in neurobiology and behavior. AIE increases adult alcohol drinking and preference, increases anxiety and reward seeking, and disrupts sleep and cognition, all risks for AUD. In addition, AIE induces changes in neuroimmune gene expression in neurons and glia that alter neurocircuitry and behavior. HMGB1 is a unique neuroimmune signal released from neurons and glia by ethanol that activates multiple proinflammatory receptors, including Toll-like receptors (TLRs), that spread proinflammatory gene induction. HMGB1 expression is increased by AIE in rat brain and in post-mortem human AUD brain, where it correlates with lifetime alcohol consumption. HMGB1 activation of TLR increase TLR expression. Human AUD brain and rat brain following AIE show increases in multiple TLRs. Brain regional differences in neurotransmitters and cell types impact ethanol responses and neuroimmune gene induction. Microglia are monocyte-like cells that provide trophic and synaptic functions, that ethanol proinflammatory signals sensitize or "prime" during repeated drinking cycles, impacting neurocircuitry. Neurocircuits are differently impacted dependent upon neuronal-glial signaling. Acetylcholine is an anti-inflammatory neurotransmitter. AIE increases HMGB1-TLR4 signaling in forebrain, reducing cholinergic neurons by silencing multiple cholinergic defining genes through upregulation of RE-1 silencing factor (REST), a transcription inhibitor known to regulate neuronal differentiation. HMGB1 REST induction reduces cholinergic neurons in basal forebrain and cholinergic innervation of hippocampus. Adult brain hippocampal neurogenesis is regulated by a neurogenic niche formed from multiple cells. In vivo AIE and in vitro studies find ethanol increases HMGB1-TLR4 signaling and other proinflammatory signaling as well as reducing trophic factors, NGF, and BDNF, coincident with loss of the cholinergic synapse marker vChAT. These changes in gene expression-transcriptomes result in reduced adult neurogenesis. Excitingly, HMGB1 antagonists, anti-inflammatories, and epigenetic modifiers like histone deacetylase inhibitors restore trophic the neurogenesis. These findings suggest anti-inflammatory and epigenetic drugs should be considered for AUD therapy and may provide long-lasting reversal of psychopathology.

2.
Alcohol Clin Exp Res (Hoboken) ; 47(3): 470-485, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36799290

RESUMEN

BACKGROUND: Adolescent intermittent ethanol (AIE) exposure causes long-term changes in the brain and behavior of adult male rodents, including persistent induction of innate immune pathways, reductions in hippocampal neurogenic and forebrain cholinergic neuronal markers, and reversal learning deficits. The current study tests the hypothesis that proinflammatory induction mediates AIE-induced (1) loss of adult neurogenesis (i.e., doublecortin (DCX) expressing immature neurons), (2) reductions in forebrain and hippocampal cholinergic markers, and (3) reversal learning deficits. METHODS: Male and female rats underwent AIE (5.0 g/kg/day ethanol or water, i.g., 2 day-on/2 day-off from postnatal day (PND) 25-54), followed by a 2-week regimen of the anti-inflammatory compound indomethacin (4.0 g/kg/day, PND 56-69) or vehicle, after which one cohort was euthanized for immunohistochemical markers (PND 70) and the second underwent the Morris water maze to assess reversal learning. RESULTS: AIE reduced adult (PND 70) DCX+ immunoreactivity (IR) and increased hippocampal expression of the innate immune signal's high-mobility group box protein 1 (HMGB1 + IR) and cyclooxygenase-2 (COX-2 + IR) in adult male and female rats. AIE also reduced choline acetyltransferase (ChAT+IR) in the basal forebrain and co-labeling of hippocampal vesicular acetylcholine transporter (VAChT+) cholinergic terminals on DCX + IR neurons. Indomethacin treatment after AIE restored molecular endpoints to control levels and rescued AIE-induced reversal learning deficits in the Morris water maze in both sexes. Of note, indomethacin produced several adverse effects selectively in control conditions, highlighting the uniquely beneficial effect of indomethacin in AIE rats. CONCLUSIONS: These data suggest that in males and females, (1) AIE persistent neuroimmune induction mediates both the loss of adult hippocampal DCX and loss of basal forebrain cholinergic neurons and their innervation to hippocampal targets, and (2) anti-inflammatory indomethacin treatment following AIE that restores these persistent molecular pathologies also restores spatial reversal learning deficits.


Asunto(s)
Etanol , Indometacina , Ratas , Animales , Masculino , Femenino , Etanol/farmacología , Indometacina/farmacología , Indometacina/metabolismo , Aprendizaje Inverso , Hipocampo , Prosencéfalo , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/patología , Neurogénesis , Colinérgicos/metabolismo , Colinérgicos/farmacología , Inmunidad Innata , Aprendizaje por Laberinto
3.
Pharmacol Rev ; 75(2): 380-396, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36781218

RESUMEN

Studies universally find early age of drinking onset is linked to lifelong risks of alcohol problems and alcohol use disorder (AUD). Assessment of the lasting effect of drinking during adolescent development in humans is confounded by the diversity of environmental and genetic factors that affect adolescent development, including emerging personality disorders and progressive increases in drinking trajectories into adulthood. Preclinical studies using an adolescent intermittent ethanol (AIE) exposure rat model of underage binge drinking avoid the human confounds and support lifelong changes that increase risks. AIE increases adult alcohol drinking, risky decision-making, reward-seeking, and anxiety as well as reductions in executive function that all increase risks for the development of an AUD. AIE causes persistent increases in brain neuroimmune signaling high-mobility group box 1 (HMGB1), Toll-like receptor, receptor for advanced glycation end products, and innate immune genes that are also found to be increased in human AUD brain. HMGB1 is released from cells by ethanol, both free and within extracellular vesicles, that act on neurons and glia, shifting transcription and cellular phenotype. AIE-induced decreases in adult hippocampal neurogenesis and loss of basal forebrain cholinergic neurons are reviewed as examples of persistent AIE-induced pathology. Both are prevented and reversed by anti-inflammatory and epigenetic drugs. Findings suggest AIE-increased HMGB1 signaling induces the RE-1 silencing transcript blunting cholinergic gene expression, shifting neuronal phenotype. Inhibition of HMGB1 neuroimmune signaling, histone methylation enzymes, and galantamine, the cholinesterase inhibitor, both prevent and reverse AIE pathology. These findings provide new targets that may reverse AUD neuropathology as well as other brain diseases linked to neuroimmune signaling. SIGNIFICANCE STATEMENT: Adolescent underage binge drinking studies find that earlier adolescent drinking is associated with lifelong alcohol problems including high levels of lifetime alcohol use disorder (AUD). Preclinical studies find the underage binge drinking adolescent intermittent ethanol (AIE) model causes lasting changes in adults that increase risks of developing adult alcohol problems. Loss of hippocampal neurogenesis and loss of basal forebrain cholinergic neurons provide examples of how AIE-induced epigenetic and neuroimmune signaling provide novel therapeutic targets for adult AUD.


Asunto(s)
Alcoholismo , Consumo Excesivo de Bebidas Alcohólicas , Proteína HMGB1 , Consumo de Alcohol en Menores , Adolescente , Animales , Humanos , Ratas , Consumo de Bebidas Alcohólicas , Alcoholismo/tratamiento farmacológico , Alcoholismo/genética , Alcoholismo/patología , Consumo Excesivo de Bebidas Alcohólicas/genética , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Consumo Excesivo de Bebidas Alcohólicas/patología , Epigénesis Genética , Etanol/efectos adversos , Proteína HMGB1/genética , Proteína HMGB1/metabolismo
4.
Alcohol Clin Exp Res ; 46(5): 759-769, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35307830

RESUMEN

BACKGROUND: Binge alcohol exposure during adolescence results in long-lasting alterations in the brain and behavior. For example, adolescent intermittent ethanol (AIE) exposure in rodents results in long-term loss of functional connectivity among prefrontal cortex (PFC) and striatal regions as well as a variety of neurochemical, molecular, and epigenetic alterations. Interneurons in the PFC and striatum play critical roles in behavioral flexibility and functional connectivity. For example, parvalbumin (PV) interneurons are known to contribute to neural synchrony and cholinergic interneurons contribute to strategy selection. Furthermore, extracellular perineuronal nets (PNNs) that surround some interneurons, particularly PV+ interneurons, further regulate cellular plasticity. The effect of AIE exposure on the expression of these markers within the PFC is not well understood. METHODS: The present study tested the hypothesis that AIE exposure reduces the expression of PV+ and choline acetyltransferase (ChAT)+ interneurons in the adult PFC and striatum and increases the related expression of PNNs (marked by binding of Wisteria floribunda agglutinin lectin) in adulthood. Male rats were exposed to AIE (5 g/kg/day, 2-days-on/2-days-off, i.e., P25 to P54) or water (CON), and brain tissue was harvested in adulthood (>P80). Immunohistochemistry and co-immunofluorescence were used to assess the expression of ChAT, PV, and PNNs within the adult PFC and striatum following AIE exposure. RESULTS: ChAT and PV interneuron densities in the striatum and PFC were unchanged after AIE exposure. However, PNN density in the PFC of AIE-exposed rats was greater than in CON rats. Moreover, significantly more PV neurons were surrounded by PNNs in AIE-exposed subjects than controls in both PFC subregions assessed: orbitofrontal cortex (CON = 34%; AIE = 40%) and medial PFC (CON = 10%; AIE = 14%). CONCLUSIONS: These findings indicate that, following AIE exposure, PV interneuron expression in the adult PFC and striatum is unaltered, while PNNs surrounding these neurons are increased. This increase in PNNs may restrict the plasticity of the ensheathed neurons, thereby contributing to impaired microcircuitry in frontostriatal connectivity and related behavioral impairments.


Asunto(s)
Etanol , Interneuronas , Adolescente , Adulto , Animales , Etanol/metabolismo , Matriz Extracelular/metabolismo , Humanos , Interneuronas/metabolismo , Masculino , Parvalbúminas/metabolismo , Corteza Prefrontal/metabolismo , Ratas
5.
Front Pharmacol ; 13: 849997, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35308225

RESUMEN

Alcohol (ethanol) use and misuse is a costly societal issue that can affect an individual across the lifespan. Alcohol use and misuse typically initiates during adolescence and generally continues into adulthood. Not only is alcohol the most widely abused drug by adolescents, but it is also one of the most widely abused drugs in the world. In fact, high rates of maternal drinking make developmental ethanol exposure the most preventable cause of neurological deficits in the Western world. Preclinical studies have determined that one of the most consistent effects of ethanol is its disruption of hippocampal neurogenesis. However, the severity, persistence, and reversibility of ethanol's effects on hippocampal neurogenesis are dependent on developmental stage of exposure and age at assessment. Complicating the neurodevelopmental effects of ethanol is the concurrent development and maturation of neuromodulatory systems which regulate neurogenesis, particularly the cholinergic system. Cholinergic signaling in the hippocampus directly regulates hippocampal neurogenesis through muscarinic and nicotinic receptor actions and indirectly regulates neurogenesis by providing anti-inflammatory regulatory control over the hippocampal environmental milieu. Therefore, this review aims to evaluate how shifting maturational patterns of the cholinergic system and its regulation of neuroimmune signaling impact ethanol's effects on adult neurogenesis. For example, perinatal ethanol exposure decreases basal forebrain cholinergic neuron populations, resulting in long-term developmental disruptions to the hippocampus that persist into adulthood. Exaggerated neuroimmune responses and disruptions in adult hippocampal neurogenesis are evident after environmental, developmental, and pharmacological challenges, suggesting that perinatal ethanol exposure induces neurogenic deficits in adulthood that can be unmasked under conditions that strain neural and immune function. Similarly, adolescent ethanol exposure persistently decreases basal forebrain cholinergic neuron populations, increases hippocampal neuroimmune gene expression, and decreases hippocampal neurogenesis in adulthood. The effects of neither perinatal nor adolescent ethanol are mitigated by abstinence whereas adult ethanol exposure-induced reductions in hippocampal neurogenesis are restored following abstinence, suggesting that ethanol-induced alterations in neurogenesis and reversibility are dependent upon the developmental period. Thus, the focus of this review is an examination of how ethanol exposure across critical developmental periods disrupts maturation of cholinergic and neuroinflammatory systems to differentially affect hippocampal neurogenesis in adulthood.

6.
Int Rev Neurobiol ; 160: 305-340, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34696877

RESUMEN

Alcohol drinking is often initiated during adolescence, and this frequently escalates to binge drinking. As adolescence is also a period of dynamic neurodevelopment, preclinical evidence has highlighted that some of the consequences of binge drinking can be long lasting with deficits persisting into adulthood in a variety of cognitive-behavioral tasks. However, while the majority of preclinical work to date has been performed in male rodents, the rapid increase in binge drinking in adolescent female humans has re-emphasized the importance of addressing alcohol effects in the context of sex as a biological variable. Here we review several of the consequences of adolescent ethanol exposure in light of sex as a critical biological variable. While some alcohol-induced outcomes, such as non-social approach/avoidance behavior and sleep disruption, are generally consistent across sex, others are variable across sex, such as alcohol drinking, sensitivity to ethanol, social anxiety-like behavior, and induction of proinflammatory markers.


Asunto(s)
Consumo de Bebidas Alcohólicas , Etanol , Consumo de Bebidas Alcohólicas/efectos adversos , Consumo de Bebidas Alcohólicas/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Etanol/toxicidad , Femenino , Masculino , Roedores , Factores Sexuales
7.
Int Rev Neurobiol ; 160: 117-173, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34696872

RESUMEN

Cognitive flexibility in decision making depends on prefrontal cortical function and is used by individuals to adapt to environmental changes in circumstances. Cognitive flexibility can be measured in the laboratory using a variety of discrete, translational tasks, including those that involve reversal learning and/or set-shifting ability. Distinct components of flexible behavior rely upon overlapping brain circuits, including different prefrontal substructures that have separable impacts on decision making. Cognitive flexibility is impaired after chronic alcohol exposure, particularly during development when the brain undergoes rapid maturation. This review examines how cognitive flexibility, as indexed by reversal and set-shifting tasks, is impacted by chronic alcohol exposure in adulthood, adolescent, and prenatal periods in humans and animal models. We also discuss areas for future study, including mechanisms that may contribute to the persistence of cognitive deficits after developmental alcohol exposure and the compacting consequences from exposure across multiple critical periods.


Asunto(s)
Trastornos del Conocimiento , Etanol , Animales , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/fisiopatología , Etanol/toxicidad , Humanos
8.
J Neuroinflammation ; 18(1): 212, 2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34530858

RESUMEN

BACKGROUND: Binge ethanol exposure during adolescence reduces hippocampal neurogenesis, a reduction which persists throughout adulthood despite abstinence. This loss of neurogenesis, indicated by reduced doublecortin+ immunoreactivity (DCX+IR), is paralleled by an increase in hippocampal proinflammatory signaling cascades. As galantamine, a cholinesterase inhibitor, has anti-inflammatory actions, we tested the hypothesis that galantamine would prevent (study 1) or restore (study 2) AIE induction of proinflammatory signals within the hippocampus as well as AIE-induced loss of hippocampal neurogenesis. METHODS: Galantamine (4 mg/kg) or vehicle (saline) was administered to Wistar rats during adolescent intermittent ethanol (AIE; 5.0 g/kg ethanol, 2 days on/2 days off, postnatal day [P] 25-54) (study 1, prevention) or after AIE during abstinent maturation to adulthood (study 2, restoration). RESULTS: Results indicate AIE reduced DCX+IR and induced cleaved caspase3 (Casp3) in DCX-expressing immature neurons. Excitingly, AIE induction of activated Casp3 in DCX-expressing neurons is both prevented and reversed by galantamine treatment, which also resulted in prevention and restoration of neurogenesis (DCX+IR). Similarly, galantamine prevented and/or reversed AIE induction of proinflammatory markers, including the chemokine (C-C motif) ligand 2 (CCL2), cyclooxygenase-2 (COX-2), and high mobility group box 1 (HMGB1) protein, suggesting that AIE induction of proinflammatory signaling mediates both cell death cascades and hippocampal neurogenesis. Interestingly, galantamine treatment increased Ki67+IR generally as well as increased pan-Trk expression specifically in AIE-treated rats but failed to reverse AIE induction of NADPH-oxidase (gp91phox). CONCLUSIONS: Collectively, our studies suggest that (1) loss of neurogenesis after AIE is mediated by persistent induction of proinflammatory cascades which drive activation of cell death machinery in immature neurons, and (2) galantamine can prevent and restore AIE disruptions in the hippocampal environmental milieu to then prevent and restore AIE-mediated loss of neurogenesis.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/tratamiento farmacológico , Etanol/toxicidad , Galantamina/uso terapéutico , Hipocampo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuroinmunomodulación/efectos de los fármacos , Factores de Edad , Animales , Consumo Excesivo de Bebidas Alcohólicas/inmunología , Consumo Excesivo de Bebidas Alcohólicas/patología , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Femenino , Galantamina/farmacología , Hipocampo/inmunología , Hipocampo/patología , Masculino , Neurogénesis/inmunología , Neuroinmunomodulación/inmunología , Ratas , Ratas Wistar
9.
Brain Sci ; 10(11)2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33126417

RESUMEN

Binge drinking is common in adolescence. Rodent studies modeling adolescent binge drinking find persistent effects on the brain's physiology, including increased expression of neuroimmune genes, impaired neurogenesis, and changes in behavioral flexibility. This study used females and males to investigate the effects of adolescent intermittent ethanol (AIE) on a battery of behaviors assessing spatial navigation using a radial arm water maze, working memory using the Hebb-Williams maze, non-spatial long-term memory using novel object recognition, and dominance using a tube dominance test. Results indicate that AIE impairs adult acquisition in spatial navigational learning with deficits predominantly driven by females. Surprisingly, AIE slowed the transition from random to serial search strategies in both sexes, suggesting AIE impairs flexibility in problem-solving processing. In the Hebb-Williams maze working memory task, adult AIE rats exhibited deficits in problem solving, resulting in more errors across the 12 maze configurations, independent of sex. Conversely, AIE decreased dominance behaviors in female rats, and at 7 months post-alcohol, female AIE rats continued to exhibit deficits in novel object recognition. These results suggest that cognitive-behavioral alterations after adolescent binge drinking persist well into middle age, despite abstinence. Future studies should focus on intervening treatment strategies in both females and males.

10.
Curr Opin Pharmacol ; 50: 9-16, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31778865

RESUMEN

Alcohol abuse and binge drinking are common during adolescence - a maturational period characterized by heightened hippocampal neuroplasticity and neurogenesis. Preclinical rodent models of adolescent binge drinking (i.e., adolescent intermittent ethanol [AIE]) find unique vulnerability of adolescent hippocampal neurogenesis with reductions persisting into adulthood after ethanol cessation. Recent discoveries implicate increased neuroimmune signaling and decreased neurotrophic support through epigenetic mechanisms in the persistent AIE-induced loss of neurogenesis. Importantly, interventions aimed at rectifying the increased neuroimmune signaling and neurotrophic-epigenetic modifications through physical activity, anti-inflammatory drugs, and histone deacetylase inhibitors protect and recover the loss of neurogenesis and cognitive deficits. The mechanisms underlying the persistent AIE-induced loss of adult hippocampal neurogenesis could contribute to broader neurodegeneration, loss of hippocampal neuroplasticity, and cognitive dysfunction.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas , Disfunción Cognitiva , Epigénesis Genética , Hipocampo/crecimiento & desarrollo , Neurogénesis , Consumo de Alcohol en Menores , Adolescente , Animales , Hipocampo/inmunología , Humanos
11.
Exp Neurol ; 318: 71-77, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31028829

RESUMEN

In the periphery insulin plays a critical role in the regulation of metabolic homeostasis by stimulating glucose uptake into peripheral organs. In the central nervous system (CNS), insulin plays a critical role in the formation of neural circuits and synaptic connections from the earliest stages of development and facilitates and promotes neuroplasticity in the adult brain. Beyond these physiological roles of insulin, a shared feature between the periphery and CNS is that decreases in insulin receptor activity and signaling (i.e. insulin resistance) contributes to the pathological consequences of type 2 diabetes (T2DM) and obesity. Indeed, clinical and preclinical studies illustrate that CNS insulin resistance elicits neuroplasticity deficits that lead to decreases in cognitive function and increased risk of neuropsychiatric disorders. The goals of this review are to provide an overview of the literature that have identified the neuroplasticity deficits observed in T2DM and obesity, as well as to discuss the potential causes and consequences of insulin resistance in the CNS, with a particular focus on how insulin resistance impacts hippocampal neuroplasticity. Interestingly, studies that have examined the effects of hippocampal-specific insulin resistance illustrate that brain insulin resistance may impair neuroplasticity independent of peripheral insulin resistance, thereby supporting the concept that restoration of brain insulin activity is an attractive therapeutic strategy to ameliorate or reverse cognitive decline observed in patients with CNS insulin resistance such as T2DM and Alzheimer's Disease.


Asunto(s)
Hipocampo/fisiopatología , Resistencia a la Insulina/fisiología , Plasticidad Neuronal/fisiología , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Hipocampo/metabolismo , Humanos
12.
Front Neuroendocrinol ; 49: 31-42, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29258741

RESUMEN

The development of the organism is a critical variable which influences the magnitude, duration, and reversibility of the effects of chronic stress. Such factors are relevant to the prefrontal cortex (PFC), as this brain region is the last to mature, the first to decline, and is highly stress-sensitive. Therefore, this review will examine the intersection between the nervous system and immune system at glutamatergic synapses in the PFC across three developmental periods: adolescence, adulthood, and aging. Glutamatergic synapses are tightly juxtaposed with microglia and astrocytes, and each of these cell types exhibits their own developmental trajectory. Not only does chronic stress differentially impact each of these cell types across development, but chronic stress also alters intercellular communication within this quad-partite synapse. These observations suggest that developmental shifts in both neural and immune function across neurons, microglia, and astrocytes mediate shifting effects of chronic stress on glutamatergic transmission.


Asunto(s)
Factores de Edad , Envejecimiento , Enfermedad Crónica , Corteza Prefrontal , Estrés Psicológico , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Femenino , Humanos , Masculino , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/inmunología , Corteza Prefrontal/metabolismo , Estrés Psicológico/inmunología , Estrés Psicológico/metabolismo
13.
Behav Brain Res ; 332: 259-268, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28600000

RESUMEN

Fetal Alcohol Syndrome (FAS) is associated with high rates of drug addiction in adulthood. One possible basis for increased drug use in this population is altered sensitivity to drug-associated contexts. This experiment utilized a rat model of FASD to examine behavioral and neural changes in the processing of drug cues in adulthood. Alcohol was given by intragastric intubation to pregnant rats throughout gestation and to rat pups during the early postnatal period (ET group). Controls consisted of a non-treated group (NC) and a pair-fed group given the intubation procedure without alcohol (IC). On postnatal day (PD) 90, rats from all treatment groups were given saline, 0.3mg/kg, 3.0mg/kg, or 10.0mg/kg cocaine pairings with a specific context in the conditioned place preference (CPP) paradigm. While control animals of both sexes showed cocaine CPP at the 3.0 and 10.0mg/kg doses, ET females also showed cocaine CPP at 0.3mg/kg. This was accompanied by a decrease in c-Fos/GAD67 cells in the nucleus accumbens (NAc) shell and GAD67-only cells in the NAc shell and PFC at this 0.3mg/kg dose. ET males failed to show cocaine CPP at the 3.0mg/kg dose. This was associated with an increase in c-Fos only-labeled cells in the NAc core and PFC at this 3.0mg/kg dose. These results suggest that developmental alcohol exposure has a sexually-dimorphic effect on cocaine's conditioning effects in adulthood and the NAc.


Asunto(s)
Cocaína/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Trastornos del Espectro Alcohólico Fetal/fisiopatología , Caracteres Sexuales , Conducta Espacial/efectos de los fármacos , Animales , Depresores del Sistema Nervioso Central/toxicidad , Condicionamiento Psicológico/fisiología , Relación Dosis-Respuesta a Droga , Etanol/toxicidad , Trastornos del Espectro Alcohólico Fetal/patología , Trastornos del Espectro Alcohólico Fetal/psicología , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/patología , Neuronas GABAérgicas/fisiología , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/patología , Núcleo Accumbens/fisiopatología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , Corteza Prefrontal/fisiopatología , Ratas Long-Evans , Conducta Espacial/fisiología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/patología , Área Tegmental Ventral/fisiopatología
14.
Physiol Behav ; 176: 207-213, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28267584

RESUMEN

The adipocyte-derived hormone leptin is an important regulator of body weight and metabolism through activation of brain leptin receptors expressed in regions such as the hypothalamus. Beyond these well described and characterized activities of leptin in the hypothalamus, it is becoming increasingly clear that the central activities of leptin extend to the hippocampus. Indeed, leptin receptors are expressed in the hippocampus where these receptors are proposed to mediate various aspects of hippocampal synaptic plasticity that ultimately impact cognitive function. This concept is supported by studies demonstrating that leptin promotes hippocampal-dependent learning and memory, as well as studies indicating that leptin resistance is associated with deficits in hippocampal-dependent behaviors and in the induction of depressive-like behaviors. The effects of leptin on cognitive/behavioral plasticity in the hippocampus may be regulated by direct activation of leptin receptors expressed in the hippocampus; additionally, leptin-mediated activation of synaptic networks that project to the hippocampus may also impact hippocampal-mediated behaviors. In view of these previous observations, the goal of this review will be to discuss the mechanisms through which leptin facilitates cognition and behavior, as well as to dissect the loci at which leptin resistance leads to impairments in hippocampal synaptic plasticity, including the development of cognitive deficits and increased risk of depressive illness in metabolic disorders such as obesity and type 2 diabetes mellitus (T2DM).


Asunto(s)
Hipocampo/metabolismo , Leptina/metabolismo , Trastornos Mentales/patología , Animales , Humanos , Leptina/uso terapéutico , Trastornos Mentales/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
15.
Neuroscience ; 350: 124-132, 2017 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-28344067

RESUMEN

Aging is associated with changes in numerous homeostatic functions, such as food intake, that are thought to be mediated by the hypothalamus. Orexin/hypocretin neurons of the hypothalamus regulate several physiological functions, including feeding, sleep and wakefulness. Evidence from both clinical and animal studies supports the notion that aging is associated with loss or dysregulation of the orexin system. Here, we used virus-mediated gene transfer to manipulate expression of orexin peptides in young and aged rats and examined behavioral and neurochemical correlates of food intake in these animals. Aged rats showed slower feeding latencies when presented with palatable food compared to young control rats, and these deficits were ameliorated by upregulation of orexin expression. Similarly, young animals treated with a virus designed to decrease preproorexin expression showed longer feeding latencies reminiscent of aged control rats. Feeding was also associated with increased acetylcholine, glutamate and GABA efflux in insular cortex of young control animals. Orexin upregulation did not restore deficits in feeding-elicited release of these neurotransmitters in aged rats, but did enhance basal neurotransmitter levels which may have contributed to the behavioral correlates of these genetic manipulations. These studies demonstrate that age-related deficits in behavioral and neurochemical measures of feeding are likely to be mediated, in part, by the orexin system. Because these same neurotransmitter systems have been shown to underlie orexin effects on cognition, treatments which increase orexin function may have potential for improving both physiological and cognitive manifestations of certain age-related disorders.


Asunto(s)
Corteza Cerebral , Conducta Alimentaria/fisiología , Neuronas/metabolismo , Orexinas/metabolismo , Transmisión Sináptica/fisiología , Envejecimiento , Animales , Corteza Cerebral/metabolismo , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Ratas Endogámicas F344 , Regulación hacia Arriba
16.
Neurosci Biobehav Rev ; 71: 267-280, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27593444

RESUMEN

Although the primary role for the immune system is to respond to pathogens, more recently, the immune system has been demonstrated to have a critical role in signaling developmental events. Of particular interest for this review is how immunocompetent microglia and astrocytes interact with glutamatergic systems to influence the development of neural circuits in the prefrontal cortex (PFC). Microglia are the resident macrophages of the brain, and astrocytes mediate both glutamatergic uptake and coordinate with microglia to respond to the general excitatory state of the brain. Cross-talk between microglia, astrocytes, and glutamatergic neurons forms a quad-partite synapse, and this review argues that interactions within this synapse have critical implications for the maturation of PFC-dependent cognitive function. Similarly, understanding developmental shifts in immune signaling may help elucidate variations in sensitivities to developmental disruptions.


Asunto(s)
Corteza Prefrontal , Astrocitos , Ácido Glutámico , Humanos , Sinapsis
17.
Physiol Behav ; 128: 126-32, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24518861

RESUMEN

Obesity-induced changes in the metabolic and endocrine milieu elicit deficits in neuroplasticity, including increased risk for development of neuropsychiatric disorders such as depressive illness. We previously demonstrated that downregulation of hypothalamic insulin receptors (hypo-IRAS) elicits a phenotype that is consistent with features of the metabolic syndrome (MetS) and that rats with this phenotype exhibit deficits in neuronal plasticity, including depressive-like behaviors such as anhedonia. Since food restriction paradigms effectively inhibit obesity-induced neuroplasticity deficits, the aim of the current study was to determine whether food restriction could reverse obesity-induced anhedonia in hypo-IRAS rats. Compared to hypo-IRAS rats provided ad lib food access, food restriction paradigms that were initiated either prior to increases in body weight or following development of the MetS/obesity phenotype effectively restored sucrose intake in hypo-IRAS rats. Moreover, food restriction paradigms were able to prevent and reverse the changes in the endocrine/metabolic/inflammatory milieu observed in hypo-IRAS, such as increases in plasma leptin and triglyceride levels and increases in pro-inflammatory cytokines such as IL-1α, IL-6 and C-reactive protein (CRP). Collectively, these results demonstrate that obesity-induced anhedonia is a reversible process and identify some potential mechanistic mediators that may be responsible for co-morbid depression in obesity.


Asunto(s)
Anhedonia/fisiología , Privación de Alimentos/fisiología , Obesidad/complicaciones , Animales , Peso Corporal/fisiología , Proteína C-Reactiva/análisis , Proteína C-Reactiva/fisiología , Citocinas/sangre , Citocinas/fisiología , Ingestión de Alimentos/fisiología , Hipotálamo/fisiología , Masculino , Obesidad/fisiopatología , Obesidad/psicología , Ratas , Ratas Sprague-Dawley , Receptor de Insulina/fisiología
18.
Physiol Behav ; 104(2): 235-41, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21036186

RESUMEN

Epidemiological studies estimate that greater than 60% of the adult US population may be categorized as either overweight or obese and there is a growing appreciation that obesity affects the functional integrity of the central nervous system (CNS). We recently developed a lentivirus (LV) vector that produces an insulin receptor (IR) antisense RNA sequence (IRAS) that when injected into the hypothalamus selectively decreases IR signaling in hypothalamus, resulting in increased body weight, peripheral adiposity and plasma leptin levels. To test the hypothesis that this obesity/hyperleptinemic phenotype would impair hippocampal synaptic transmission, we examined short term potentiation (STP) and long term potentiation (LTP) in the hippocampus of rats that received the LV-IRAS construct or the LV-Control construct in the hypothalamus (hypo-IRAS and hypo-Con, respectively). Stimulation of the Schaffer collaterals elicits STP that develops into LTP in the CA1 region of hypo-Con rats; conversely, hypo-IRAS rats exhibit STP that fails to develop into LTP. To more closely examine the potential role of hyperleptinemia in these electrophysiological deficits, hypo-IRAS were subjected to mild food restriction paradigms that would either: 1) prevent the development of the obesity phenotype; or 2) reverse an established obesity phenotype in hypo-IRAS rats. Both of these paradigms restored LTP in the CA1 region and reversed the decreases in the phosphorylated/total ratio of GluA1 Ser845 AMPA receptor subunit expression observed in the hippocampus of hypo-IRAS rats. Collectively, these data support the hypothesis that obesity impairs hippocampal synaptic transmission and support the hypothesis that these deficits are mediated through the impairment of hippocampal leptin activity.


Asunto(s)
Privación de Alimentos/fisiología , Hipocampo/fisiopatología , Leptina/metabolismo , Potenciación a Largo Plazo/fisiología , Obesidad/patología , Adiposidad/fisiología , Animales , Área Bajo la Curva , Autorradiografía , Peso Corporal/fisiología , Corticosterona/sangre , Modelos Animales de Enfermedad , Regulación hacia Abajo/fisiología , Ensayo de Inmunoadsorción Enzimática/métodos , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Técnicas In Vitro , Insulina/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Fosforilación/efectos de los fármacos , ARN sin Sentido/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptor de Insulina/genética , Receptores AMPA/metabolismo , Serina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...