Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 3021, 2024 02 06.
Article in English | MEDLINE | ID: mdl-38321123

ABSTRACT

The initiation of alcohol use early in life is one of the strongest predictors of developing a future alcohol use disorder. Clinical studies have identified specific behaviors during early childhood that predict an increased risk for excess alcohol consumption later in life. These behaviors, including increased hyperactivity, anxiety, novelty-seeking, exploratory behavior, impulsivity, and alcohol-seeking, are similarly stimulated in children and adolescent offspring of mothers who drink alcohol during pregnancy. Here we tested larval zebrafish in addition to young pre-weanling rats and found this repertoire of early behaviors along with the overconsumption of alcohol during adolescence to be increased by embryonic ethanol exposure. With hypocretin/orexin (Hcrt) neurons known to be stimulated by ethanol and involved in mediating these alcohol-related behaviors, we tested their function in larval zebrafish and found optogenetic activation of Hcrt neurons to stimulate these same early alcohol-related behaviors and later alcohol intake, suggesting that these neurons have an important role in producing these behaviors. Together, these results show zebrafish to be an especially useful animal model for investigating the diverse neuronal systems mediating behavioral changes at young ages that are produced by embryonic ethanol exposure and predict an increased risk for developing alcohol use disorder.


Subject(s)
Alcoholism , Ethanol , Child, Preschool , Humans , Pregnancy , Female , Child , Animals , Rats , Adolescent , Orexins/genetics , Zebrafish , Optogenetics , Alcohol Drinking , Neurons
2.
Cells ; 12(10)2023 05 16.
Article in English | MEDLINE | ID: mdl-37408233

ABSTRACT

Studies in zebrafish and rats show that embryonic ethanol exposure at low-moderate concentrations stimulates hypothalamic neurons expressing hypocretin/orexin (Hcrt) that promote alcohol consumption, effects possibly involving the chemokine Cxcl12 and its receptor Cxcr4. Our recent studies in zebrafish of Hcrt neurons in the anterior hypothalamus (AH) demonstrate that ethanol exposure has anatomically specific effects on Hcrt subpopulations, increasing their number in the anterior AH (aAH) but not posterior AH (pAH), and causes the most anterior aAH neurons to become ectopically expressed further anterior in the preoptic area (POA). Using tools of genetic overexpression and knockdown, our goal here was to determine whether Cxcl12a has an important function in mediating the specific effects of ethanol on these Hcrt subpopulations and their projections. The results demonstrate that the overexpression of Cxcl12a has stimulatory effects similar to ethanol on the number of aAH and ectopic POA Hcrt neurons and the long anterior projections from ectopic POA neurons and posterior projections from pAH neurons. They also demonstrate that knockdown of Cxcl12a blocks these effects of ethanol on the Hcrt subpopulations and projections, providing evidence supporting a direct role of this specific chemokine in mediating ethanol's stimulatory effects on embryonic development of the Hcrt system.


Subject(s)
Chemokines , Ethanol , Zebrafish , Animals , Embryonic Development , Ethanol/pharmacology , Neurons/physiology , Orexins
3.
Sci Rep ; 13(1): 8448, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37231149

ABSTRACT

Numerous studies in animals demonstrate that embryonic exposure to ethanol (EtOH) at low-moderate doses stimulates neurogenesis and increases the number of hypothalamic neurons expressing the peptide, hypocretin/orexin (Hcrt). A recent study in zebrafish showed that this effect on the Hcrt neurons in the anterior hypothalamus (AH) is area specific, evident in the anterior (aAH) but not posterior (pAH) part of this region. To understand specific factors that may determine the differential sensitivity to EtOH of these Hcrt subpopulations, we performed additional measures in zebrafish of their cell proliferation, co-expression of the opioid dynorphin (Dyn), and neuronal projections. In association with the increase in Hcrt neurons in the aAH but not pAH, EtOH significantly increased only in the aAH the proliferation of Hcrt neurons and their number lacking Dyn co-expression. The projections of these subpopulations differed markedly in their directionality, with those from the pAH primarily descending to the locus coeruleus and those from the aAH ascending to the subpallium, and they were both stimulated by EtOH, which induced specifically the most anterior subpallium-projecting Hcrt neurons to become ectopically expressed beyond the aAH. These differences between the Hcrt subpopulations suggest they are functionally distinct in their regulation of behavior.


Subject(s)
Dynorphins , Zebrafish , Animals , Orexins , Ethanol/toxicity , Neurons/physiology , Cell Proliferation
4.
Sci Rep ; 13(1): 1447, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36702854

ABSTRACT

Embryonic ethanol exposure in zebrafish and rats, while stimulating hypothalamic hypocretin/orexin (Hcrt) neurons along with alcohol consumption and related behaviors, increases the chemokine receptor Cxcr4 that promotes neuronal migration and may mediate ethanol's effects on neuronal development. Here we performed a more detailed anatomical analysis in zebrafish of ethanol's effects on the Cxcl12a/Cxcr4b system throughout the entire brain as it relates to Hcrt neurons developing within the anterior hypothalamus (AH) where they are normally located. We found that ethanol increased these Hcrt neurons only in the anterior part of the AH and induced ectopic Hcrt neurons further anterior in the preoptic area, and these effects along with ethanol-induced behaviors were completely blocked by a Cxcr4 antagonist. Analysis of cxcl12a transcripts and internalized Cxcr4b receptors throughout the brain showed they both exhibited natural posterior-to-anterior concentration gradients, with levels lowest in the posterior AH and highest in the anterior telencephalon. While stimulating their density in all areas and maintaining these gradients, ethanol increased chemokine expression only in the more anterior and ectopic Hcrt neurons, effects blocked by the Cxcr4 antagonist. These findings demonstrate how increased chemokine expression acting along natural gradients mediates ethanol-induced anterior migration of ectopic Hcrt neurons and behavioral disturbances.


Subject(s)
Ethanol , Zebrafish , Animals , Rats , Orexins/metabolism , Zebrafish/metabolism , Ethanol/toxicity , Ethanol/metabolism , Hypothalamus, Posterior/metabolism , Chemokines/metabolism , Neurons/metabolism
5.
Addict Biol ; 27(6): e13238, 2022 11.
Article in English | MEDLINE | ID: mdl-36301208

ABSTRACT

Embryonic exposure to ethanol increases the risk for alcohol use disorder in humans and stimulates alcohol-related behaviours in different animal models. Evidence in rats and zebrafish suggests that this phenomenon induced by ethanol at low-moderate concentrations involves a stimulatory effect on neurogenesis and density of hypothalamic neurons expressing the peptides, hypocretin/orexin (Hcrt) and melanin-concentrating hormone (MCH), known to promote alcohol consumption. Building on our report in zebrafish showing that ethanol induces ectopic expression of Hcrt neurons outside the hypothalamus, we investigated here whether embryonic ethanol exposure also induces ectopic peptide neurons in rats similar to zebrafish and affects their morphological characteristics and if these ectopic neurons are functional and have a role in the ethanol-induced disturbances in behaviour. We demonstrate in rats that ethanol at a low-moderate dose, in addition to increasing Hcrt and MCH neurons in the lateral hypothalamus where they are normally concentrated, induces ectopic expression of these peptide neurons further anterior in the nucleus accumbens core and ventromedial caudate putamen where they have not been previously observed and causes morphological changes relative to normally located hypothalamic neurons. Similar to rats, embryonic ethanol exposure at a low-moderate dose in zebrafish induces ectopic Hcrt neurons anterior to the hypothalamus and alters their morphology. Notably, laser ablation of these ectopic Hcrt neurons blocks the behavioural effects induced by ethanol exposure, including increased anxiety and locomotor activity. These findings suggest that the ectopic peptide neurons are functional and contribute to the ethanol-induced behavioural disturbances related to the overconsumption of alcohol.


Subject(s)
Ethanol , Neurons , Orexins , Prenatal Exposure Delayed Effects , Animals , Rats , Ethanol/metabolism , Hypothalamus/drug effects , Neurons/drug effects , Orexins/metabolism , Zebrafish
6.
Article in English | MEDLINE | ID: mdl-35176416

ABSTRACT

Prenatal alcohol exposure (PAE) increases alcohol consumption and risk for alcohol use disorder. This phenomenon in rodents is suggested to involve a stimulatory effect of PAE, in female more than male offspring, on neurogenesis and density of neurons expressing neuropeptides in lateral hypothalamus (LH), including melanin-concentrating hormone (MCH), known to promote alcohol intake. With evidence suggesting a role for fibroblast growth factor 2 (FGF2) and its receptor FGFR1 in stimulating neurogenesis and alcohol drinking, we investigated here whether the FGF2-FGFR1 system is involved in the PAE-induced increase in MCH neurons, in postnatal offspring of pregnant rats given ethanol orally (embryonic day 10-15) at a low-moderate (2 g/kg/day) or high (5 g/kg/day) dose. Our results demonstrate that PAE at the low-moderate but not high dose stimulates FGF2 and FGFR1 gene expression and increases the density of MCH neurons co-expressing FGF2, only in females, but FGFR1 in both sexes. PAE induces this effect in the dorsal but not ventral area of the LH. Further analysis of FGF2 and FGFR1 transcripts within individual MCH neurons reveals an intracellular, sex-dependent effect, with PAE increasing FGF2 transcripts positively related to FGFR1 in the nucleus as well as cytoplasm of females but transcripts only in the cytoplasm of males. Peripheral injection of FGF2 itself (80 µg/kg, s.c.) in pregnant rats mimics these effects of PAE. Together, these results support the involvement of the FGF2-FGFR1 system in mediating the PAE-induced, sex dependent increase in density of MCH neurons, possibly contributing to increased alcohol consumption in the offspring.


Subject(s)
Fibroblast Growth Factor 2 , Prenatal Exposure Delayed Effects , Animals , Ethanol , Female , Fibroblast Growth Factor 2/adverse effects , Fibroblast Growth Factor 2/metabolism , Male , Neurons , Peptides/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Sprague-Dawley
7.
J Neuroinflammation ; 17(1): 207, 2020 Jul 10.
Article in English | MEDLINE | ID: mdl-32650794

ABSTRACT

BACKGROUND: Clinical and animal studies show that alcohol consumption during pregnancy produces lasting behavioral disturbances in offspring, including increased alcohol drinking, which are linked to inflammation in the brain and disturbances in neurochemical systems that promote these behaviors. These include the neuropeptide, melanin-concentrating hormone (MCH), which is mostly expressed in the lateral hypothalamus (LH). Maternal ethanol administration at low-to-moderate doses, while stimulating MCH neurons without affecting apoptosis or gliogenesis, increases in LH the density of neurons expressing the inflammatory chemokine C-C motif ligand 2 (CCL2) and its receptor CCR2 and their colocalization with MCH. These neural effects associated with behavioral changes are reproduced by maternal CCL2 administration, reversed by a CCR2 antagonist, and consistently stronger in females than males. The present study investigates in the embryo the developmental origins of this CCL2/CCR2-mediated stimulatory effect of maternal ethanol exposure on MCH neurons. METHODS: Pregnant rats from embryonic day 10 (E10) to E15 during peak neurogenesis were orally administered ethanol at a moderate dose (2 g/kg/day) or peripherally injected with CCL2 or CCR2 antagonist to test this neuroimmune system's role in ethanol's actions. Using real-time quantitative PCR, immunofluorescence histochemistry, in situ hybridization, and confocal microscopy, we examined in embryos at E19 the CCL2/CCR2 system and MCH neurons in relation to radial glia progenitor cells in the hypothalamic neuroepithelium where neurons are born and radial glia processes projecting laterally through the medial hypothalamus that provide scaffolds for neuronal migration into LH. RESULTS: We demonstrate that maternal ethanol increases radial glia cell density and their processes while stimulating the CCL2/CCR2 system and these effects are mimicked by maternal administration of CCL2 and blocked by a CCR2 antagonist. While stimulating CCL2 colocalization with radial glia and neurons but not microglia, ethanol increases MCH neuronal number near radial glia cells and making contact along their processes projecting into LH. Further tests identify the CCL2/CCR2 system in NEP as a primary source of ethanol's sexually dimorphic actions. CONCLUSIONS: These findings provide new evidence for how an inflammatory chemokine pathway functions within neuroprogenitor cells to mediate ethanol's long-lasting, stimulatory effects on peptide neurons linked to adolescent drinking behavior.


Subject(s)
Chemokine CCL2/metabolism , Ethanol/toxicity , Hypothalamus/metabolism , Neuroepithelial Cells/metabolism , Receptors, CCR2/metabolism , Sex Characteristics , Animals , Embryonic Development/drug effects , Embryonic Development/physiology , Ethanol/administration & dosage , Female , Hypothalamus/drug effects , Hypothalamus/embryology , Male , Neuroepithelial Cells/drug effects , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Stem Cells/metabolism
9.
Alcohol Clin Exp Res ; 44(4): 866-879, 2020 04.
Article in English | MEDLINE | ID: mdl-32020622

ABSTRACT

BACKGROUND: Prenatal exposure to ethanol (EtOH) has lasting effects on neuropeptide and neuroimmune systems in the brain alongside detrimental alcohol-related behaviors. At low-to-moderate doses, prenatal EtOH stimulates neurogenesis in lateral hypothalamus (LH) and increases neurons that express the orexigenic peptides hypocretin/orexin (Hcrt/OX) and melanin-concentrating hormone (MCH), and the proinflammatory chemokine CCL2, which through its receptor CCR2 stimulates cell differentiation and movement. Our recent studies demonstrated that CCL2 and CCR2 colocalize with MCH neurons and are involved in EtOH's stimulatory effect on their development but show no relation to Hcrt/OX. Here, we investigated another chemokine, CXCL12, and its receptor, CXCR4, which promote neurogenesis and neuroprogenitor cell proliferation, to determine if they also exhibit peptide specificity in their response to EtOH exposure. METHODS: Pregnant rats were intraorally administered a moderate dose of EtOH (2 g/kg/d) from embryonic day 10 (E10) to E15. Their embryos and postnatal offspring were examined using real-time quantitative PCR and immunofluorescence histochemistry, to determine if EtOH affects CXCL12 and CXCR4 and the colocalization of CXCR4 with Hcrt/OX and MCH neurons in the LH and with radial glia neuroprogenitor cells in the hypothalamic neuroepithelium (NEP). RESULTS: Prenatal EtOH strongly stimulated CXCL12 and CXCR4 in LH neurons of embryos and postnatal offspring. This stimulation was significantly stronger in Hcrt/OX than MCH neurons in LH and also occurred in radial glia neuroprogenitor cells dense in the NEP. These effects were sexually dimorphic, consistently stronger in females than males. CONCLUSIONS: While showing prenatal EtOH exposure to have a sexually dimorphic, stimulatory effect on CXCL12 and CXCR4 in LH similar to CCL2 and its receptor, these results reveal their distinct relationship to the peptide neurons, with the former closely related to Hcrt/OX and the latter to MCH, and they link EtOH's actions in LH to a stimulatory effect on neuroprogenitor cells in the NEP.


Subject(s)
Central Nervous System Depressants/pharmacology , Chemokine CXCL12/drug effects , Ependymoglial Cells/drug effects , Ethanol/pharmacology , Hypothalamic Area, Lateral/drug effects , Neural Stem Cells/drug effects , Neurons/drug effects , Receptors, CXCR4/drug effects , Animals , Animals, Newborn , Cell Proliferation/drug effects , Chemokine CXCL12/metabolism , Embryo, Mammalian , Ependymoglial Cells/metabolism , Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , Melanins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/metabolism , Orexins/metabolism , Pituitary Hormones/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/metabolism
10.
Neuroscience ; 443: 188-205, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31982472

ABSTRACT

Clinical and animal studies show maternal alcohol consumption during pregnancy causes in offspring persistent alterations in neuroimmune and neurochemical systems known to increase alcohol drinking and related behaviors. Studies in lateral hypothalamus (LH) demonstrate in adolescent offspring that maternal oral administration of ethanol stimulates the neuropeptide, melanin-concentrating hormone (MCH), together with the inflammatory chemokine C-C motif ligand 2 (CCL2) and its receptor CCR2 which are increased in most MCH neurons. These effects, consistently stronger in females than males, are detected in embryos, not only in LH but hypothalamic neuroepithelium (NEP) along the third ventricle where neurons are born and CCL2 is stimulated within radial glia progenitor cells and their laterally projecting processes that facilitate MCH neuronal migration toward LH. With ethanol's effects similarly produced by maternal peripheral CCL2 administration and blocked by CCR2 antagonist, we tested here using in utero intracerebroventricular (ICV) injections whether CCL2 acts locally within the embryonic NEP. After ICV injection of CCL2 (0.1 µg/µl) on embryonic day 14 (E14) when neurogenesis peaks, we observed in embryos just before birth (E19) a significant increase in endogenous CCL2 within radial glia cells and their processes in NEP. These auto-regulatory effects, evident only in female embryos, were accompanied by increased density of CCL2 and MCH neurons in LH, more strongly in females than males. These results support involvement of embryonic CCL2/CCR2 neuroimmune system in radial glia progenitor cells in mediating sexually dimorphic effects of maternal challenges such as ethanol on LH MCH neurons that colocalize CCL2 and CCR2.


Subject(s)
Hypothalamic Hormones , Third Ventricle , Animals , Chemokine CCL2/metabolism , Ependymoglial Cells/metabolism , Female , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Male , Neurons/metabolism , Peptides , Pituitary Hormones , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, CCR2/metabolism , Stem Cells/metabolism , Third Ventricle/metabolism
11.
Neuroscience ; 424: 155-171, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31705896

ABSTRACT

Maternal consumption of ethanol during pregnancy is known to increase the offspring's risk for developing alcohol use disorders and associated behavioral disturbances. Studies in adolescent and adult animals suggest the involvement of neuroimmune and neurochemical systems in the brain that control these behaviors. To understand the origin of these effects during early developmental stages, we examined in the embryo and neonate the effects of maternal intraoral administration of ethanol (2 g/kg/day) from embryonic day 10 (E10) to E15 on the inflammatory chemokine C-C motif ligand 2 (CCL2) and its receptor CCR2 in a specific, dense population of neurons in the lateral hypothalamus (LH), where they are closely related to an orexigenic neuropeptide, melanin-concentrating hormone (MCH), known to promote ethanol consumption and related behaviors. We found that prenatal ethanol exposure increases the expression and density of CCL2 and CCR2 cells along with MCH neurons in the LH and the colocalization of CCL2 with MCH. We also discovered that these effects are sexually dimorphic, consistently stronger in female embryos, and are blocked by maternal administration of a CCL2 antibody (1 and 5 µg/day, i.p., E10-E15) that neutralizes endogenous CCL2 and of a CCR2 antagonist INCB3344 (1 mg/day, i.p., E10-E15) that blocks CCL2's main receptor. These results, which in the embryo anatomically and functionally link the CCL2/CCR2 system to MCH neurons in the LH, suggest an important role for this neuroimmune system in mediating ethanol's sexually dimorphic, stimulatory effect on MCH neurons that may promote higher level of alcohol consumption described in females.


Subject(s)
Chemokine CCL2/biosynthesis , Ethanol/administration & dosage , Hypothalamus/metabolism , Prenatal Exposure Delayed Effects/metabolism , Receptors, CCR2/biosynthesis , Sex Characteristics , Animals , Animals, Newborn , Chemokine CCL2/antagonists & inhibitors , Ethanol/toxicity , Female , Hypothalamus/drug effects , Hypothalamus/embryology , Male , Neurons/drug effects , Neurons/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Pyrrolidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, CCR2/antagonists & inhibitors
12.
J Neurosci ; 38(42): 9072-9090, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30201767

ABSTRACT

Clinical and animal studies show that ethanol exposure and inflammation during pregnancy cause similar behavioral disturbances in the offspring. While ethanol is shown to stimulate both neuroimmune and neurochemical systems in adults, little is known about their anatomical relationship in response to ethanol in utero and whether neuroimmune factors mediate ethanol's effects on neuronal development and behavior in offspring. Here we examined in female and male adolescent rats a specific population of neurons concentrated in lateral hypothalamus, which coexpress the inflammatory chemokine C-C motif ligand 2 (CCL2) or its receptor CCR2 with the orexigenic neuropeptide, melanin-concentrating hormone (MCH), that promotes ethanol drinking behavior. We demonstrate that maternal administration of ethanol (2 g/kg/d) from embryonic day 10 (E10) to E15, while having little impact on glia, stimulates expression of neuronal CCL2 and CCR2, increases density of both large CCL2 neurons colocalizing MCH and small CCL2 neurons surrounding MCH neurons, and stimulates ethanol drinking and anxiety in adolescent offspring. We show that these neuronal and behavioral changes are similarly produced by maternal administration of CCL2 (4 or 8 µg/kg/d, E10-E15) and blocked by maternal administration of a CCR2 antagonist INCB3344 (1 mg/kg/d, E10-E15), and these effects of ethanol and CCL2 are sexually dimorphic, consistently stronger in females. These results suggest that this neuronal CCL2/CCR2 system closely linked to MCH neurons has a role in mediating the effects of maternal ethanol exposure on adolescent offspring and contributes to the higher levels of adolescent risk factors for alcohol use disorders described in women.SIGNIFICANCE STATEMENT Ethanol consumption and inflammatory agents during pregnancy similarly increase alcohol intake and anxiety in adolescent offspring. To investigate how neurochemical and neuroimmune systems interact to mediate these disturbances, we examined a specific population of hypothalamic neurons coexpressing the inflammatory chemokine CCL2 and its receptor CCR2 with the neuropeptide, melanin-concentrating hormone. We demonstrate in adolescent offspring that maternal administration of CCL2, like ethanol, stimulates these neurons and increases ethanol drinking and anxiety, and these effects of ethanol are blocked by maternal CCR2 antagonist and consistently stronger in females. This suggests that neuronal chemokine signaling linked to neuropeptides mediates effects of maternal ethanol exposure on adolescent offspring and contributes to higher levels of adolescent risk factors for alcohol use disorders in women.


Subject(s)
Chemokine CCL2/metabolism , Ethanol/administration & dosage , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , Melanins/metabolism , Pituitary Hormones/metabolism , Prenatal Exposure Delayed Effects/metabolism , Receptors, CCR2/metabolism , Sex Characteristics , Alcohol Drinking , Animals , Anxiety/chemically induced , Behavior, Animal , Cell Count , Female , Neurons/metabolism , Pregnancy , Rats, Sprague-Dawley
13.
J Neurochem ; 135(5): 918-31, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26332891

ABSTRACT

Ingestion of a high-fat diet composed mainly of the saturated fatty acid, palmitic (PA), and the unsaturated fatty acid, oleic (OA), stimulates transcription in the brain of the opioid neuropeptide, enkephalin (ENK), which promotes intake of substances of abuse. To understand possible underlying mechanisms, this study examined the nuclear receptors, peroxisome proliferator-activated receptors (PPARs), and tested in hypothalamic and forebrain neurons from rat embryos whether PPARs regulate endogenous ENK and the fatty acids themselves affect these PPARs and ENK. The first set of experiments demonstrated that knocking down PPARδ, but not PPARα or PPARγ, increased ENK transcription, activation of PPARδ by an agonist decreased ENK levels, and PPARδ neurons coexpressed ENK, suggesting that PPARδ negatively regulates ENK. In the second set of experiments, PA treatment of hypothalamic and forebrain neurons had no effect on PPARδ protein while stimulating ENK mRNA and protein, whereas OA increased both mRNA and protein levels of PPARδ in forebrain neurons while having no effect on ENK mRNA and increasing ENK levels. These findings show that PA has a strong, stimulatory effect on ENK and weak effect on PPARδ protein, whereas OA has a strong stimulatory effect on PPARδ and weak effect on ENK, consistent with the inhibitory effect of PPARδ on ENK. They suggest a function for PPARδ, perhaps protective in nature, in embryonic neurons exposed to fatty acids from a fat-rich diet and provide evidence for a mechanism contributing to differential effects of saturated and monounsaturated fatty acids on neurochemical systems involved in consummatory behavior. Our findings show that PPARδ in forebrain and hypothalamic neurons negatively regulates enkephalin (ENK), a peptide known to promote ingestive behavior. This inverse relationship is consistent with our additional findings, that a saturated (palmitic; PA) compared to a monounsaturated fatty acid (oleic; OA) has a strong stimulatory effect on ENK and weak effect on PPARδ. These results suggest that PPARδ protects against the neuronal effects of fatty acids, which differentially affect neurochemical systems involved in ingestive behavior.


Subject(s)
Enkephalins/metabolism , Fatty Acids/metabolism , Hypothalamus/cytology , Neurons/metabolism , PPAR delta/metabolism , Prosencephalon/cytology , Animals , Cells, Cultured , Diet, High-Fat , Dose-Response Relationship, Drug , Embryo, Mammalian , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Oleic Acid/pharmacology , Palmitic Acid/pharmacology , RNA, Messenger , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Thiazoles/pharmacology , Transfection
14.
Alcohol ; 49(5): 479-89, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25979531

ABSTRACT

Clinical studies demonstrate frequent co-existence of nicotine and alcohol abuse and suggest that this may result, in part, from the ready access to and intake of fat-rich diets. Whereas animal studies show that high-fat diet intake in adults can enhance the consumption of either nicotine or ethanol and that maternal consumption of a fat-rich diet during pregnancy increases operant responding for nicotine in offspring, little is known about the impact of dietary fat on the co-abuse of these two drugs. The goal of this study was to test in Long-Evans rats the effects of perinatal exposure to fat on the co-use of nicotine and ethanol, using a novel paradigm that involves simultaneous intravenous (IV) self-administration of these two drugs. Fat- vs. chow-exposed offspring were characterized and compared, first in terms of their nicotine self-administration behavior, then in terms of their nicotine/ethanol self-administration behavior, and lastly in terms of their self-administration of ethanol in the absence of nicotine. The results demonstrate that maternal consumption of fat compared to low-fat chow during gestation and lactation significantly stimulates nicotine self-administration during fixed-ratio testing. It also increases nicotine/ethanol self-administration during fixed-ratio and dose-response testing, with BEC elevated to 120 mg/dL, and causes an increase in breakpoint during progressive ratio testing. Of particular note is the finding that rats perinatally exposed to fat self-administer significantly more of the nicotine/ethanol mixture as compared to nicotine alone, an effect not evident in the chow-control rats. After removal of nicotine from the nicotine/ethanol mixture, this difference between the fat- and chow-exposed rats was lost, with both groups failing to acquire the self-administration of ethanol alone. Together, these findings suggest that perinatal exposure to a fat-rich diet, in addition to stimulating self-administration of nicotine, causes an even greater vulnerability to the excessive co-use of nicotine and ethanol.


Subject(s)
Diet, High-Fat , Dietary Fats/pharmacology , Ethanol/administration & dosage , Ethanol/pharmacology , Nicotine/administration & dosage , Nicotine/pharmacology , Prenatal Exposure Delayed Effects/psychology , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Ethanol/blood , Female , Male , Pregnancy , Rats , Rats, Long-Evans , Reinforcement Schedule , Self Administration
15.
J Neurosci ; 33(34): 13600-11, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23966683

ABSTRACT

Animal and clinical studies show that gestational exposure to nicotine increases the propensity of offspring to consume nicotine, but the precise mechanism mediating this behavioral phenomenon is unclear. The present study in Sprague Dawley rats examined the possibility that the orexigenic peptide systems, enkephalin (ENK) and orexin (OX), which are stimulated by nicotine in adult animals and promote consummatory behavior, may be similarly responsive to nicotine's stimulatory effect in utero while having long-term behavioral consequences. The results demonstrated that nicotine exposure during gestation at low doses (0.75 or 1.5 mg/kg/d) significantly increased mRNA levels and density of neurons that express ENK in the hypothalamic paraventricular nucleus and central nucleus of the amygdala, OX, and another orexigenic peptide, melanin-concentrating hormone, in the perifornical lateral hypothalamus in preweanling offspring. These effects persisted in the absence of nicotine, at least until puberty. Colabeling of the cell proliferation marker BrdU with the neuronal marker NeuN and peptides revealed a marked stimulatory effect of prenatal nicotine on neurogenesis, but not gliogenesis, and also on the number of newly generated neurons expressing ENK, OX, or melanin-concentrating hormone. During adolescence, offspring also exhibited significant behavioral changes, increased consumption of nicotine and other substances of abuse, ethanol and a fat-rich diet, with no changes in chow and water intake or body weight. These findings reveal a marked sensitivity during gestation of the orexigenic peptide neurons to low nicotine doses that may increase the offspring's propensity to overconsume substances of abuse during adolescence.


Subject(s)
Amygdala/cytology , Gene Expression Regulation, Developmental/drug effects , Hypothalamus/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Neurogenesis/drug effects , Neuropeptides/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Prenatal Exposure Delayed Effects/chemically induced , Age Factors , Amygdala/drug effects , Amygdala/growth & development , Amygdala/metabolism , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Enkephalins/genetics , Enkephalins/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Hypothalamic Hormones/genetics , Hypothalamic Hormones/metabolism , Hypothalamus/drug effects , Hypothalamus/growth & development , Hypothalamus/metabolism , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/genetics , Melanins/genetics , Melanins/metabolism , Neurons/drug effects , Neurons/metabolism , Neuropeptides/genetics , Orexins , Phosphopyruvate Hydratase , Pituitary Hormones/genetics , Pituitary Hormones/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
16.
Psychopharmacology (Berl) ; 230(4): 509-24, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23836027

ABSTRACT

RATIONALE: While clinical studies show maternal consumption of palatable fat-rich diets during pregnancy to negatively impact the children's behaviors and increase their vulnerability to drug abuse, the precise behavioral and neurochemical mechanisms mediating these phenomena have yet to be examined. OBJECTIVE: The study examined in rats whether gestational exposure to a high-fat diet (HFD) can increase the offspring's propensity to use nicotine and whether disturbances in central nicotinic cholinergic signaling accompany this behavioral effect. METHODS: Rat offspring exposed perinatally to a HFD or chow diet were characterized in terms of their nicotine self-administration behavior in a series of operant response experiments and the activity of acetylcholinesterase (AChE) and density of nicotinic ACh receptors (nAChRs) in different brain areas. RESULT: Perinatal HFD compared to chow exposure increased nicotine-self administration behavior during fixed ratio and dose-response testing and caused an increase in breakpoint using progressive ratio testing, while nicotine seeking in response to nicotine prime-induced reinstatement was reduced. This behavioral change induced by the HFD was associated with a significant reduction in activity of AChE in the midbrain, hypothalamus, and striatum and increased density of ß2-nAChRs in the ventral tegmental area and substantia nigra and of α7-nAChRs in the lateral and ventromedial hypothalamus. CONCLUSIONS: Perinatal exposure to a HFD increases the vulnerability of the offspring to excessive nicotine use by enhancing its reward potential, and these behavioral changes are accompanied by a stimulation of nicotinic cholinergic signaling in mesostriatal and hypothalamic brain areas important for reinforcement and consummatory behavior.


Subject(s)
Diet, High-Fat , Nicotine/administration & dosage , Prenatal Exposure Delayed Effects , Reward , Acetylcholinesterase/metabolism , Animals , Behavior, Animal/physiology , Brain/metabolism , Conditioning, Operant/physiology , Female , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism , Reinforcement Schedule , Self Administration , Signal Transduction/physiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
17.
Alcohol ; 46(6): 559-68, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22703995

ABSTRACT

The process of ethanol anticipation is a particularly important phenomenon that can determine subsequent drug-taking behavior. Recent studies suggest that systems within the medial prefrontal cortex (mPFC), during anticipation, may contribute to the goal-directed seeking of ethanol. The current investigation examined the possibility that the opioid peptide enkephalin (ENK), known to mediate some of the reinforcing properties of ethanol, may function in the mPFC during the anticipation of ethanol access. Using a limited access (3 h/d) paradigm for 10 days with 20% ethanol, Sprague-Dawley rats were first identified either as low drinkers (LD, <1.0 g/kg/3 h) or as high drinkers (HD, >2.0 g/kg/3 h) that exhibited a long-term phenotype of high ethanol consumption and a significant ethanol deprivation effect. During the anticipation period immediately preceding daily ethanol access, the HD rats compared to LD or Control animals with ad libitum ethanol access exhibited increased anticipatory behaviors, including greater exploratory behavior in a novel open field as revealed by significantly more time spent in the rearing position (+53-65%, p < 0.05) and increased number of rears made (+33-44%, p < 0.05) and greater novelty-seeking behavior in a hole-board apparatus revealed by an increase in total (+50-52%, p < 0.05) and novel nose pokes (+45-48%, p < 0.05). In the HD rats, analysis of the mPFC using real-time quantitative PCR showed significantly greater mRNA levels of ENK (p < 0.05) and the mu-opioid receptor (MOR) (p < 0.05), but not delta-opioid receptor (DOR), and this increase in ENK expression was found, using in situ hybridization, to occur specifically in the prelimbic (PrL) subregion of the mPFC. When injected into the PrL during the anticipation period, a MOR agonist but not DOR agonist significantly increased consumption of 20% ethanol (p < 0.05). These findings support the role of ENK, acting through MOR within the PrL to promote the anticipation and excessive consumption of ethanol.


Subject(s)
Alcohol Drinking/metabolism , Behavior, Animal/physiology , Enkephalins/biosynthesis , Exploratory Behavior/physiology , Animals , Anticipation, Psychological/physiology , Behavior, Animal/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, Methionine/analogs & derivatives , Enkephalin, Methionine/pharmacology , Enkephalins/genetics , Ethanol/metabolism , Exploratory Behavior/drug effects , Male , Prefrontal Cortex/physiology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics
18.
Regul Pept ; 173(1-3): 13-20, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-21903140

ABSTRACT

While a high-fat diet when compared to low-fat diet is known to produce overeating and health complications, less is known about the effects produced by fat-rich diets differing in their specific composition of fat. This study examined the effects of a high-fat diet containing relatively high levels of saturated compared to unsaturated fatty acids (HiSat) to a high-fat diet with higher levels of unsaturated fatty acids (USat). A HiSat compared to USat meal caused rats to consume more calories in a subsequent chow test meal. The HiSat meal also increased circulating levels of triglycerides (TG) and expression of the orexigenic peptides, galanin (GAL) in the hypothalamic paraventricular nucleus (PVN) and orexin (OX) in the perifornical lateral hypothalamus (PFLH). A similar increase in TG levels and PVN GAL and PFLH OX was also seen in rats given chronic access to the HiSat compared to USat diet, while neuropeptide Y (NPY) and agouti-related protein (AgRP) in the arcuate nucleus showed decreased expression. The importance of TG in producing these changes was supported by the finding that the TG-lowering medication gemfibrozil as compared to vehicle, when peripherally administered before consumption of a HiSat meal, significantly decreased the expression of OX, while increasing the expression of NPY and AgRP. These findings substantiate the importance of the fat composition in a diet, indicating that those rich in saturated compared to unsaturated fatty acids may promote overeating by increasing circulating lipids and specific hypothalamic peptides, GAL and OX, known to preferentially stimulate the consumption of a fat-rich diet.


Subject(s)
Dietary Fats/pharmacology , Eating/drug effects , Fatty Acids/pharmacology , Hypothalamic Hormones/metabolism , Triglycerides/blood , Animals , Appetite , Diet, High-Fat , Female , Gemfibrozil/pharmacology , Gene Expression/drug effects , Hypolipidemic Agents/pharmacology , Hypothalamic Hormones/genetics , Hypothalamus/metabolism , Male , Rats , Rats, Sprague-Dawley
19.
Alcohol ; 44(4): 323-34, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20692550

ABSTRACT

To investigate mechanisms in outbred animals that increase the propensity to consume ethanol, it is important to identify and characterize these animals before or at early stages in their exposure to ethanol. In the present study, different measures were examined in adult Sprague-Dawley rats to determine whether they can predict long-term propensity to overconsume ethanol. Before consuming 9% ethanol with a two-bottle choice paradigm, rats were examined with the commonly used behavioral measures of novelty-induced locomotor activity and anxiety, as assessed during 15 min in an open-field activity chamber. Two additional measures, intake of a low 2% ethanol concentration or circulating triglyceride (TG) levels after a meal, were also examined with respect to their ability to predict chronic 9% ethanol consumption. The results revealed significant positive correlations across individual rats between the amount of 9% ethanol ultimately consumed and three of these different measures, with high scores for activity, 2% ethanol intake, and TGs identifying rats that consume 150% more ethanol than rats with low scores. Measurements of hypothalamic peptides that stimulate ethanol intake suggest that they contribute early to the greater ethanol consumption predicted by these high scores. Rats with high 2% ethanol intake or high TGs, two measures found to be closely related, had significantly elevated expression of enkephalin (ENK) and galanin (GAL) in the hypothalamic paraventricular nucleus (PVN) but no change in neuropeptide Y (NPY) in the arcuate nucleus (ARC). This is in contrast to rats with high activity scores, which in addition to elevated PVN ENK expression showed enhanced NPY in the ARC but no change in GAL. Elevated ENK is a common characteristic related to all three predictors of chronic ethanol intake, whereas the other peptides differentiate these predictors, with GAL enhanced with high 2% ethanol intake and TG measures but NPY related to activity.


Subject(s)
Alcohol Drinking/metabolism , Enkephalins/biosynthesis , Ethanol/administration & dosage , Galanin/biosynthesis , Hypothalamus/metabolism , Neuropeptide Y/biosynthesis , Peptides/physiology , Animals , Hypothalamus/drug effects , Male , Motor Activity/drug effects , Motor Activity/physiology , Peptides/metabolism , Predictive Value of Tests , Rats , Rats, Sprague-Dawley
20.
Pharmacol Biochem Behav ; 96(4): 413-22, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20600243

ABSTRACT

The goal of this study is to examine the expression pattern of orexigenic peptides, orexin (OX) and melanin-concentrating hormone (MCH), in the perifornical lateral hypothalamus (PFLH) in subpopulations of Sprague-Dawley rats differing in their propensity to overconsume a high-fat diet. Immediately after an initial 5-day screening test that predicts long-term consumption, rats identified as high-fat consumers (HFC), ingesting 35% more calories of a high-fat relative to low-fat chow diet, had significantly elevated mRNA expression of OX in the perifornical but not lateral hypothalamic area and of MCH mRNA in both areas, when compared to control rats that consume similar amounts of these diets. This same OX and MCH expression pattern was seen in HFC rats maintained for two weeks on a low-fat chow diet, indicating that increased expression of these orexigenic peptides, occurring independently of the high-fat diet, may be an inherent characteristic of these rats. These HFC rats were also more active and slightly more anxious than controls, as measured by line crossings and time spent in the periphery or middle segments of an open field. Together, these results demonstrate that animals prone to overeating a high-fat diet show a baseline increase in orexigenic peptide expression in the PFLH along with higher behavioral arousal, which together may contribute to their increased consummatory behavior.


Subject(s)
Dietary Fats/administration & dosage , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Melanins/metabolism , Neuropeptides/metabolism , Pituitary Hormones/metabolism , Animals , Base Sequence , DNA Primers , Hypothalamic Hormones/genetics , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/genetics , Male , Melanins/genetics , Neuropeptides/genetics , Orexins , Pituitary Hormones/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...