Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 38
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Neurotoxicol Teratol ; 102: 107341, 2024.
Article de Anglais | MEDLINE | ID: mdl-38490565

RÉSUMÉ

Prenatal exposure to alcohol or cannabinoids can produce enduring neurobiological, cognitive, and behavioral changes in the offspring. Furthermore, prenatal co-exposure to alcohol and cannabinoids induces malformations in brain regions associated with reward and stress-related circuitry. This study examined the effects of co-exposure to alcohol and the synthetic cannabinoid (SCB) CP55,940 throughout gastrulation and neurulation in rats on basal corticosterone levels and a battery of behavioral tests during adolescence and alcohol self-administration in adulthood. Importantly, we find that prenatal alcohol exposure (PAE) caused lower baseline corticosterone levels in adolescent males and females. Co-exposure to alcohol + CP produced hyperactivity during open field test in males, but not females. During the two-bottle choice alcohol-drinking procedure, prenatal cannabinoid exposed male and female adolescent rats drank more alcohol than their vehicle-exposed controls. In adulthood, female rats treated with prenatal cannabinoid exposure (PCE), showed an overall total increase in alcohol intake during alcohol self-administration; but this was not found in males. When the reinforcer was changed to a 1% sucrose solution, male rats exposed to PCE, showed a reduced self-administration compared to vehicle-exposed males, potentially indicative of an anhedonic response. This lower self-administration persisted when 20% alcohol was reintroduced to the sucrose solution. Lastly, following an abstinence period, there were no changes due to prenatal drug exposure in either males or females. Overall, these data suggest lasting consequences of prenatal alcohol and cannabinoid exposure during adolescence and adulthood in male and female rats.


Sujet(s)
Cannabinoïdes , Effets différés de l'exposition prénatale à des facteurs de risque , Humains , Rats , Femelle , Mâle , Animaux , Grossesse , Corticostérone , Effets différés de l'exposition prénatale à des facteurs de risque/induit chimiquement , Éthanol/pharmacologie , Saccharose
2.
Birth Defects Res ; 116(1): e2292, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38116840

RÉSUMÉ

BACKGROUND: Prenatal alcohol exposure during gastrulation (embryonic day [E] 7 in mice, ~3rd week of human pregnancy) impairs eye, facial, and cortical development, recapitulating birth defects characteristic of Fetal Alcohol Syndrome (FAS). However, it is not known whether the prevalence or severity of craniofacial features associated with FAS is affected by biological sex. METHODS: The current study administered either alcohol (2.9 g/kg, two i.p. doses, 4 hr apart) or vehicle to pregnant C57BL/6J females on E7, prior to gonadal sex differentiation, and assessed fetal morphology at E17. RESULTS: Whereas sex did not affect fetal size in controls, alcohol-exposed females were smaller than both control females and alcohol-treated males. Alcohol exposure increased the incidence of eye defects to a similar degree in males and females. Together, these data suggest that females might be more sensitive to the general developmental effects of alcohol, but not effects specific to the craniofacies. Whole transcriptomic analysis of untreated E7 embryos found 214 differentially expressed genes in females vs. males, including those in pathways related to cilia and mitochondria, histone demethylase activity, and pluripotency. CONCLUSION: Gastrulation-stage alcohol induces craniofacial malformations in male and female mouse fetuses at similar rates and severity, though growth deficits are more prevalent females. These findings support the investigation of biological sex as a contributing factor in prenatal alcohol studies.


Sujet(s)
Malformations crâniofaciales , Troubles du spectre de l'alcoolisation foetale , Effets différés de l'exposition prénatale à des facteurs de risque , Humains , Femelle , Mâle , Grossesse , Animaux , Souris , Gastrulation , Souris de lignée C57BL , Effets différés de l'exposition prénatale à des facteurs de risque/étiologie , Éthanol/effets indésirables , Troubles du spectre de l'alcoolisation foetale/génétique , Malformations crâniofaciales/induit chimiquement
3.
bioRxiv ; 2023 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-37873078

RÉSUMÉ

Prenatal exposure to alcohol or cannabinoids can produce enduring neurobiological, cognitive, and behavioral changes in the offspring. Furthermore, prenatal co-exposure to alcohol and cannabinoids induces malformations in brain regions associated with reward and stress-related circuitry. This study examined the effects of co-exposure to alcohol and the synthetic cannabinoid (SCB) CP55,940 throughout gastrulation and neurulation in rats on basal corticosterone levels and a battery of behavioral tests during adolescence and alcohol self-administration in adulthood. Importantly, we find that prenatal alcohol exposure (PAE) caused lower baseline corticosterone levels in adolescent males and females. Co-exposure to alcohol + CP produced hyperactivity during open field test in males, but not females. During the two-bottle choice alcohol-drinking procedure, prenatal cannabinoid exposed male and female adolescent rats drank more alcohol than their vehicle-exposed controls. In adulthood, female rats treated with prenatal cannabinoid exposure (PCE), showed an overall total increase in alcohol intake during alcohol self-administration; but this was not found in males. When the reinforcer was changed to a 1% sucrose solution, male rats exposed to PCE, showed a reduced self-administration compared to vehicle-exposed males, potentially indicative of an anhedonic response. This lower self-administration persisted when 20% alcohol was reintroduced to the sucrose solution. Lastly, following an abstinence period, there were no changes due to prenatal drug exposure in either males or females. Overall, these data suggest lasting consequences of prenatal alcohol and cannabinoid exposure during adolescence and adulthood in male and female rats.

4.
Birth Defects Res ; 114(19): 1229-1243, 2022 11 15.
Article de Anglais | MEDLINE | ID: mdl-35396933

RÉSUMÉ

BACKGROUND: During early development, alcohol exposure causes apoptotic cell death in discrete regions of the embryo which are associated with distinctive patterns of later-life abnormalities. In gastrulation, which occurs during the third week of human pregnancy, alcohol targets the ectoderm, the precursor of the eyes, face, and brain. This midline tissue loss leads to the craniofacial dysmorphologies, such as microphthalmia and a smooth philtrum, which define fetal alcohol syndrome (FAS). An important regulator of alcohol-induced cell death is the pro-apoptotic protein Bax. The current study determines if mice lacking the Bax gene are less susceptible to the pathogenic effects of gastrulation-stage alcohol exposure. METHODS: Male and female Bax+/- mice mated to produce embryos with full (-/- ) or partial (+/- ) Bax deletions, or Bax+/+ wild-type controls. On Gestational Day 7 (GD 7), embryos received two alcohol (2.9 g/kg, 4 hr apart), or control exposures. A subset of embryos was collected 12 hr later and examined for the presence of apoptotic cell death, while others were examined on GD 17 for the presence of FAS-like facial features. RESULTS: Full Bax deletion reduced embryonic apoptotic cell death and the incidence of fetal eye and face malformations, indicating that Bax normally facilitates the development of alcohol-induced defects. An RNA-seq analysis of GD 7 Bax+/+ and Bax-/- embryos revealed 63 differentially expressed genes, some of which may interact with the Bax deletion to further protect against apoptosis. CONCLUSIONS: Overall, these experiments identify that Bax is a primary teratogenic mechanism of gastrulation-stage alcohol exposure.


Sujet(s)
Troubles du spectre de l'alcoolisation foetale , Gastrulation , Protéine Bax , Animaux , Femelle , Humains , Mâle , Souris , Grossesse , Protéine Bax/métabolisme , Éthanol/effets indésirables , Troubles du spectre de l'alcoolisation foetale/anatomopathologie , Exposition maternelle
5.
J Neurosci Res ; 100(8): 1585-1601, 2022 08.
Article de Anglais | MEDLINE | ID: mdl-35014067

RÉSUMÉ

Ethanol exposure during the early stages of embryonic development can lead to a range of morphological and behavioral differences termed fetal alcohol spectrum disorders (FASDs). In a zebrafish model, we have shown that acute ethanol exposure at 8-10 hr postfertilization (hpf), a critical time of development, produces birth defects similar to those clinically characterized in FASD. Dysregulation of the Sonic hedgehog (Shh) pathway has been implicated as a molecular basis for many of the birth defects caused by prenatal alcohol exposure. We observed in zebrafish embryos that shh expression was significantly decreased by ethanol exposure at 8-10 hpf, while smo expression was much less affected. Treatment of zebrafish embryos with SAG or purmorphamine, small molecule Smoothened agonists that activate Shh signaling, ameliorated the severity of ethanol-induced developmental malformations including altered eye size and midline brain development. Furthermore, this rescue effect of Smo activation was dose dependent and occurred primarily when treatment was given after ethanol exposure. Markers of Shh signaling (gli1/2) and eye development (pax6a) were restored in embryos treated with SAG post-ethanol exposure. Since embryonic ethanol exposure has been shown to produce later-life neurobehavioral impairments, juvenile zebrafish were examined in the novel tank diving test. Our results further demonstrated that in zebrafish embryos exposed to ethanol, SAG treatment was able to mitigate long-term neurodevelopmental impairments related to anxiety and risk-taking behavior. Our results indicate that pharmacological activation of the Shh pathway at specific developmental timing markedly diminishes the severity of alcohol-induced birth defects.


Sujet(s)
Troubles du spectre de l'alcoolisation foetale , Effets différés de l'exposition prénatale à des facteurs de risque , Animaux , Embryon non mammalien/métabolisme , Éthanol/toxicité , Femelle , Troubles du spectre de l'alcoolisation foetale/traitement médicamenteux , Troubles du spectre de l'alcoolisation foetale/métabolisme , Protéines Hedgehog/métabolisme , Humains , Grossesse , Danio zébré/métabolisme
6.
Alcohol Clin Exp Res ; 45(10): 1965-1979, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34581462

RÉSUMÉ

BACKGROUND: Alcohol exposure during the gastrulation stage of development causes the craniofacial and brain malformations that define fetal alcohol syndrome. These malformations, such as a deficient philtrum, are exemplified by a loss of midline tissue and correspond, at least in part, to regionally selective cell death in the embryo. The tumor suppressor protein Tp53 is an important mechanism for cell death, but the role of Tp53 in the consequences of alcohol exposure during the gastrulation stage has yet to be examined. The current studies used mice and zebrafish to test whether genetic loss of Tp53 is a conserved mechanism to protect against the effects of early developmental stage alcohol exposure. METHODS: Female mice, heterozygous for a mutation in the Tp53 gene, were mated with Tp53 heterozygous males, and the resulting embryos were exposed during gastrulation on gestational day 7 (GD 7) to alcohol (two maternal injections of 2.9 g/kg, i.p., 4 h apart) or a vehicle control. Zebrafish mutants or heterozygotes for the tp53zdf1  M214K mutation and their wild-type controls were exposed to alcohol (1.5% or 2%) beginning 6 h postfertilization (hpf), the onset of gastrulation. RESULTS: Examination of GD 17 mice revealed that eye defects were the most common phenotype among alcohol-exposed fetuses, occurring in nearly 75% of the alcohol-exposed wild-type fetuses. Tp53 gene deletion reduced the incidence of eye defects in both the heterozygous and mutant fetuses (to about 35% and 20% of fetuses, respectively) and completely protected against alcohol-induced facial malformations. Zebrafish (4 days postfertilization) also demonstrated alcohol-induced reductions of eye size and trabeculae length that were less common and less severe in tp53 mutants, indicating a protective effect of tp53 deletion. CONCLUSIONS: These results identify an evolutionarily conserved role of Tp53 as a pathogenic mechanism for alcohol-induced teratogenesis.


Sujet(s)
Malformations dues aux médicaments et aux drogues/étiologie , Malformations crâniofaciales/étiologie , Éthanol/effets indésirables , Troubles du spectre de l'alcoolisation foetale/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Malformations dues aux médicaments et aux drogues/métabolisme , Animaux , Malformations crâniofaciales/métabolisme , Femelle , Mâle , Souris , Grossesse , Tératogenèse , Danio zébré
7.
Reprod Toxicol ; 105: 136-147, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34492310

RÉSUMÉ

Neurulation-stage alcohol exposure (NAE; embryonic day [E] 8-10) is associated with midline craniofacial and CNS defects that likely arise from disruption of morphogen pathways, such as Sonic hedgehog (Shh). Notably, midline anomalies are also a hallmark of genetic ciliopathies such as Joubert syndrome. We tested whether NAE alters Shh pathway signaling and the number and function of primary cilia, organelles critical for Shh pathway transduction. Female C57BL/6 J mice were administered two doses of alcohol (2.9 g/kg/dose) or vehicle on E9. Embryos were collected 6, 12, or 24 h later, and changes to Shh, cell cycle genes, and primary cilia were measured in the rostroventral neural tube (RVNT). Within the first 24 h post-NAE, reductions in Shh pathway and cell cycle gene expression and the ratio of Gli3 forms in the full-length activator state were observed. RVNT volume and cell layer width were reduced at 12 h. In addition, altered expression of multiple cilia-related genes was observed at 6 h post-NAE. As a further test of cilia gene-ethanol interaction, mice heterozygous for Kif3a exhibited perturbed behavior during adolescence following NAE compared to vehicle-treated mice, and Kif3a heterozygosity exacerbated the hyperactive effects of NAE on exploratory activity. These data demonstrate that NAE downregulates the Shh pathway in a region of the neural tube that gives rise to alcohol-sensitive brain structures and identifies disruption of primary cilia function, or a "transient ciliopathy", as a possible cellular mechanism of prenatal alcohol pathogenesis.


Sujet(s)
Cils vibratiles/génétique , Éthanol/effets indésirables , Protéines Hedgehog/génétique , Tube neural/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/génétique , Animaux , Comportement animal , Cycle cellulaire/génétique , Femelle , Régulation de l'expression des gènes au cours du développement , Kinésine/génétique , Mâle , Échange foetomaternel , Souris de lignée C57BL , Souris transgéniques , Grossesse
8.
Sci Rep ; 9(1): 16057, 2019 11 05.
Article de Anglais | MEDLINE | ID: mdl-31690747

RÉSUMÉ

We tested whether cannabinoids (CBs) potentiate alcohol-induced birth defects in mice and zebrafish, and explored the underlying pathogenic mechanisms on Sonic Hedgehog (Shh) signaling. The CBs, Δ9-THC, cannabidiol, HU-210, and CP 55,940 caused alcohol-like effects on craniofacial and brain development, phenocopying Shh mutations. Combined exposure to even low doses of alcohol with THC, HU-210, or CP 55,940 caused a greater incidence of birth defects, particularly of the eyes, than did either treatment alone. Consistent with the hypothesis that these defects are caused by deficient Shh, we found that CBs reduced Shh signaling by inhibiting Smoothened (Smo), while Shh mRNA or a CB1 receptor antagonist attenuated CB-induced birth defects. Proximity ligation experiments identified novel CB1-Smo heteromers, suggesting allosteric CB1-Smo interactions. In addition to raising concerns about the safety of cannabinoid and alcohol exposure during early embryonic development, this study establishes a novel link between two distinct signaling pathways and has widespread implications for development, as well as diseases such as addiction and cancer.


Sujet(s)
Cannabinoïdes/toxicité , Troubles du spectre de l'alcoolisation foetale/métabolisme , Protéines Hedgehog/métabolisme , Récepteur cannabinoïde de type CB1/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tératogenèse/effets des médicaments et des substances chimiques , Animaux , Éthanol/effets indésirables , Éthanol/pharmacologie , Femelle , Troubles du spectre de l'alcoolisation foetale/anatomopathologie , Souris , Récepteur Smoothened/métabolisme
9.
PLoS One ; 13(4): e0194767, 2018.
Article de Anglais | MEDLINE | ID: mdl-29617407

RÉSUMÉ

In many experiments using fetal mice, it is necessary to determine the sex of the individual fetus. However, other than genotyping for sex-specific genes, there is no convenient, reliable method of sexing mice between gestational day (GD) 16.5 and GD 18.0. We designed a rapid, relatively simple visual method to determine the sex of mouse fetuses in the GD 16.5-GD 18.0 range that can be performed as part of a routine morphological assessment. By examining the genitalia for the presence or absence of key features, raters with minimal experience with the method were able to correctly identify the sex of embryos with 99% accuracy, while raters with no experience were 95% accurate. The critical genital features include: the presence or absence of urethral seam or proximal urethral meatus; the shape of the genitalia, and the presence or absence of an area related to the urethral plate. By comparing these morphological features of the external genitalia, we show a simple, accurate, and fast way to determine the sex of late stage mouse fetuses. Integrating this method into regular morphological assessments will facilitate the determination of sex differences in fetuses between GD 16.5 and GD 18.0.


Sujet(s)
Système génital/anatomie et histologie , Détermination du sexe/méthodes , Animaux , Femelle , Génotype , Âge gestationnel , Mâle , Souris , Souris de lignée C57BL , Photographie (méthode) , Grossesse , Facteurs de transcription SOX/génétique , Facteurs de transcription SOX/métabolisme
10.
Birth Defects Res ; 109(11): 860-865, 2017 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-28504423

RÉSUMÉ

BACKGROUND: Genetic factors influence the physical and neurobehavioral manifestations of prenatal alcohol exposure (PAE). Animal models allow the investigation of specific genes that confer vulnerability to, or protection from, birth defects associated with fetal alcohol spectrum disorders (FASDs). The objective of the present experiments was to determine if genetic alterations in the Sonic Hedgehog (Shh) signaling pathways affect the vulnerability to PAE-induced skeletal defects involving the forelimbs and/or hindlimbs. METHOD: Wild-type C57BL/6J female mice were bred with males in which one copy of the Shh or Gli2 genes had been knocked out, to produce litters with both wild-type (+/+) and heterozygous (+/-) embryos. Alcohol doses (two injections of 2.9 g/kg, 4 hours apart) or vehicles were administered starting at gestational day (GD) 9.25, 9.5, or 9.75, a critical exposure time for inducing limb defects. Limb defects were examined at GD 17 using a dysmorphology scale based on abnormalities ranging from increased interdigital spacing to the deletion of multiple fingers and the ulna. RESULTS: Alcohol treatment caused a high incidence of forelimb defects, particularly on the right side, that was higher in Shh+/- and Gli2+/- fetuses compared to wild-type fetuses. Dysmorphology scores were also significantly higher in the Shh+/- and Gli2+/- mice. CONCLUSIONS: These results extend previous findings demonstrating enhanced sensitivity to PAE-induced craniofacial dysmorphology and support the hypothesis that genetic alterations in the Shh signaling pathway influences the vulnerability to alcohol-induced birth defects. Moreover, these results emphasize the importance of understanding the interactions between genes and prenatal exposure to alcohol or other teratogens. Birth Defects Research 109:860-865, 2017. © 2017 Wiley Periodicals, Inc.


Sujet(s)
Éthanol/effets indésirables , Protéines Hedgehog/effets des médicaments et des substances chimiques , Protéine à doigts de zinc Gli2/effets des médicaments et des substances chimiques , Animaux , Femelle , Troubles du spectre de l'alcoolisation foetale/génétique , Troubles du spectre de l'alcoolisation foetale/physiopathologie , Prédisposition génétique à une maladie , Hétérozygote , Facteurs de transcription Krüppel-like/génétique , Anomalies morphologiques congénitales des membres/complications , Anomalies morphologiques congénitales des membres/étiologie , Mâle , Souris , Souris de lignée C57BL , Mutation , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque/étiologie , Transduction du signal/effets des médicaments et des substances chimiques , Tératogènes
11.
Birth Defects Res ; 109(1): 49-54, 2017 01 20.
Article de Anglais | MEDLINE | ID: mdl-27801979

RÉSUMÉ

BACKGROUND: While pharmacological activation of the Hedgehog (HH) signaling pathway may have therapeutic benefits for developmental and adult diseases, its teratogenic potential is of concern. The membrane molecule Smoothened (SMO) transduces HH signaling and can be acutely modulated by antagonists and agonists. The objective of the current experiments was to determine how maternal treatment with the Smo agonist, SAG, affects the developing limb. METHODS: Pregnant C57BL/6J mice received a single injection of SAG (15, 17, or 20 mg/kg, i.p.) or its vehicle on gestational day (GD) 9.25, the time of limb bud induction. Embryos were examined on GD 15 for gross dysmorphology and skeletal staining was performed to visualize the number and type of digits on the fore- and hindlimbs. Additionally, in situ hybridization was performed 4 hr after GD 9.25 SAG administration to determine SAG's effects on Gli1 and Gli2 mRNA expression. RESULTS: The most prevalent effect of SAG was the dose-dependent induction of pre-axial polydactyly; defects ranged from a broad thumb to the duplication of two finger-like digits on the preaxial side of the thumb. The highest SAG dose was effective in ca. 80% of the embryos and increased Gli1 and Gli2 mRNA expression in the limb bud, with Gli1 mRNA being the most upregulated. CONCLUSION: Preaxial polydactyly can be caused in the developing embryo by acute maternal administration of a Smo agonist that activates HH signaling. These results are consistent with the preaxial polydactyly induced in developmental disorders associated with mutations in HH signaling genes.Birth Defects Research 109:49-54, 2017. © 2016 Wiley Periodicals, Inc.


Sujet(s)
Cyclohexylamines/effets indésirables , Cyclohexylamines/métabolisme , Polydactylie/physiopathologie , Thiophènes/effets indésirables , Thiophènes/métabolisme , Animaux , Membres , Femelle , Anomalies morphologiques de la main/génétique , Anomalies morphologiques de la main/métabolisme , Protéines Hedgehog/génétique , Bourgeons de membre/métabolisme , Mâle , Souris , Souris de lignée C57BL , Mutation , Polydactylie/génétique , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque , Transduction du signal/génétique , Récepteur Smoothened/agonistes , Récepteur Smoothened/métabolisme , Pouce/malformations , Pouce/physiopathologie , Facteurs de transcription/génétique , Protéine à doigt de zinc GLI1/effets des médicaments et des substances chimiques , Protéine à doigt de zinc GLI1/génétique , Protéine à doigts de zinc Gli2/effets des médicaments et des substances chimiques , Protéine à doigts de zinc Gli2/génétique
12.
Neurotoxicol Teratol ; 58: 15-22, 2016.
Article de Anglais | MEDLINE | ID: mdl-26708672

RÉSUMÉ

Potent synthetic cannabinoids (SCBs) are illegally distributed drugs of abuse that are frequently consumed in spite of their adverse consequences. This study was designed to determine if the toxicity observed in adults also extends to the prenatal period by examining the developmental toxicity/teratogenicity of one of these SCBs, CP-55,940, in a mammalian model. First, immunohistochemistry was employed for cannabinoid receptor 1 (CB1) localization within gestational day (GD) 8 mouse embryos; this receptor was identified in the cranial neural plate, suggesting that the endogenous cannabinoid system may be involved in normal development. Based on this information and on previous avian teratogenicity studies, the current investigation focused on cannabinoid exposure during neurulation. The treatment paradigm involved acute i.p. administration of vehicle, 0.0625, 0.125, 0.25, 0.5, 1.0, or 2.0mg/kg CP-55,940 to time-mated C57Bl/6J mice on their 8th day of pregnancy (n>10 litters per treatment group). On GD 17, litters were harvested and examined for numbers of live, dead, or resorbed fetuses, as well as for fetal weight, length, and gross morphological abnormalities. No effect on litter size, fetal weight, or crown rump length was seen at any of the CP-55,940 dosages tested. Major malformations involving the craniofacies and/or eyes were noted in all drug-treated groups. Selected fetuses with craniofacial malformations were histologically sectioned and stained, allowing investigation of brain anomalies. Observed craniofacial, ocular, and brain abnormalities in drug-treated fetuses included lateral and median facial clefts, cleft palate, microphthalmia, iridial coloboma, anophthalmia, exencephaly, holoprosencephaly, and cortical dysplasia. With the most commonly observed defects involving the eyes, the incidence and severity of readily identifiable ocular malformations were utilized as a basis for dose-response analyses. Ocular malformation ratings revealed dose-dependent CP-55,940 teratogenicity within the full range of dosages tested. While examination of additional critical periods and in depth mechanistic studies is warranted, the results of this investigation clearly show the dose-dependent teratogenicity of this SCB.


Sujet(s)
Malformations dues aux médicaments et aux drogues/embryologie , Encéphale/effets des médicaments et des substances chimiques , Encéphale/embryologie , Encéphale/anatomopathologie , Cyclohexanols/toxicité , Effets différés de l'exposition prénatale à des facteurs de risque/anatomopathologie , Animaux , Cyclohexanols/administration et posologie , Relation dose-effet des médicaments , Femelle , Mâle , Souris , Souris de lignée C57BL , Neurulation/effets des médicaments et des substances chimiques , Grossesse , Récepteur cannabinoïde de type CB1/métabolisme , Tératogènes
13.
Neuropsychopharmacology ; 40(11): 2614-22, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-25881115

RÉSUMÉ

The OPRM1 A118G polymorphism is the most widely studied µ-opioid receptor (MOR) variant. Although its involvement in acute alcohol effects is well characterized, less is known about the extent to which it alters responses to opioids. Prior work has shown that both electrophysiological and analgesic responses to morphine but not to fentanyl are moderated by OPRM1 A118G variation, but the mechanism behind this dissociation is not known. Here we found that humanized mice carrying the 118GG allele (h/mOPRM1-118GG) were less sensitive than h/mOPRM1-118AA littermates to the rewarding effects of morphine and hydrocodone but not those of other opioids measured with intracranial self-stimulation. Reduced morphine reward in 118GG mice was associated with decreased dopamine release in the nucleus accumbens and reduced effects on GABA release in the ventral tegmental area that were not due to changes in drug potency or efficacy in vitro or receptor-binding affinity. Fewer MOR-binding sites were observed in h/mOPRM1-118GG mice, and pharmacological reduction of MOR availability unmasked genotypic differences in fentanyl sensitivity. These findings suggest that the OPRM1 A118G polymorphism decreases sensitivity to low-potency agonists by decreasing receptor reserve without significantly altering receptor function.


Sujet(s)
Analgésiques morphiniques/pharmacologie , Noyau accumbens/métabolisme , Récepteur mu/métabolisme , Récompense , Aire tegmentale ventrale/métabolisme , Animaux , Modèles animaux de maladie humaine , Dopamine/métabolisme , Cellules HEK293 , Humains , Mâle , Souris transgéniques , Activité motrice/effets des médicaments et des substances chimiques , Activité motrice/physiologie , Antagonistes narcotiques/pharmacologie , Noyau accumbens/effets des médicaments et des substances chimiques , Polymorphisme de nucléotide simple , Récepteur mu/génétique , Autostimulation , Techniques de culture de tissus , Aire tegmentale ventrale/effets des médicaments et des substances chimiques , Acide gamma-amino-butyrique/métabolisme
14.
Alcohol ; 49(3): 207-17, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25709101

RÉSUMÉ

The range of defects that fall within fetal alcohol spectrum disorder (FASD) includes persistent behavioral problems, with anxiety and depression being two of the more commonly reported issues. Previous studies of rodent FASD models suggest that interference with hypothalamic-pituitary-adrenal (HPA) axis structure and/or function may be the basis for some of the prenatal alcohol (ethanol) exposure (PAE)-induced behavioral abnormalities. Included among the previous investigations are those illustrating that maternal alcohol treatment limited to very early stages of pregnancy (i.e., gestational day [GD]7 in mice; equivalent to the third week post-fertilization in humans) can cause structural abnormalities in areas such as the hypothalamus, pituitary gland, and other forebrain regions integral to controlling stress and behavioral responses. The current investigation was designed to further examine the sequelae of prenatal alcohol insult at this early time period, with particular attention to HPA axis-associated functional changes in adult mice. The results of this study reveal that GD7 PAE in mice causes HPA axis dysfunction, with males and females showing elevated corticosterone (CORT) and adrenocorticotropic hormone (ACTH) levels, respectively, following a 15-min restraint stress exposure. Males also showed elevated CORT levels following an acute alcohol injection of 2.0 g/kg, while females displayed blunted ACTH levels. Furthermore, analysis showed that anxiety-like behavior was decreased after GD7 PAE in female mice, but was increased in male mice. Collectively, the results of this study show that early gestational alcohol exposure in mice alters long-term HPA axis activity and behavior in a sexually dimorphic manner.


Sujet(s)
Hormone corticotrope/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Dépresseurs du système nerveux central/pharmacologie , Corticostérone/métabolisme , Éthanol/pharmacologie , Axe hypothalamohypophysaire/effets des médicaments et des substances chimiques , Axe hypophyso-surrénalien/effets des médicaments et des substances chimiques , Hormone corticotrope/métabolisme , Animaux , Anxiété/métabolisme , Anxiété/psychologie , Hyperalcoolisation rapide , Dépression/métabolisme , Dépression/psychologie , Modèles animaux de maladie humaine , Femelle , Troubles du spectre de l'alcoolisation foetale/métabolisme , Troubles du spectre de l'alcoolisation foetale/psychologie , Axe hypothalamohypophysaire/métabolisme , Mâle , Souris , Axe hypophyso-surrénalien/métabolisme , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/psychologie , Contention physique , Facteurs sexuels , Stress psychologique/métabolisme , Stress psychologique/psychologie
15.
J Pharmacol Exp Ther ; 350(2): 322-9, 2014 Aug.
Article de Anglais | MEDLINE | ID: mdl-24917543

RÉSUMÉ

C57BL/6J (C57) and DBA/2J (DBA) mice respond differently to drugs that affect dopamine systems, including alcohol. The current study compared effects of D1 and D2 receptor agonists and antagonists, and the interaction between D1/D2 antagonists and alcohol, on intracranial self-stimulation in male C57 and DBA mice to determine the role of dopamine receptors in the effects of alcohol on brain stimulation reward (BSR). In the initial strain comparison, dose effects on BSR thresholds and maximum operant response rates were determined for the D1 receptor agonist SKF-82958 (±-6-chloro-7,8-dihydroxy-3-allyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine; 0.1-0.56 mg/kg) and antagonist SCH 23390 (+-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepinehydrochloride; 0.003-0.056 mg/kg), and the D2 receptor agonist quinpirole (0.1-3.0 mg/kg) and antagonist raclopride (0.01-0.56 mg/kg). For the alcohol interaction, SCH 23390 (0.003 mg/kg) or raclopride (0.03 mg/kg) was given before alcohol (0.6-2.4 g/kg p.o.). D1 antagonism dose-dependently elevated and SKF-82958 dose-dependently lowered BSR threshold in both strains; DBA mice were more sensitive to SKF-82958 effects. D2 antagonism dose-dependently elevated BSR threshold only in C57 mice. Low doses of quinpirole elevated BSR threshold equally in both strains, whereas higher doses of quinpirole lowered BSR threshold only in C57 mice. SCH 23390, but not raclopride, prevented lowering of BSR threshold by alcohol in DBA mice. Conversely, raclopride, but not SCH 23390, prevented alcohol potentiation of BSR in C57 mice. These results extend C57 and DBA strain differences to D1/D2 sensitivity of BSR, and suggest differential involvement of D1 and D2 receptors in the acute rewarding effects of alcohol in these two mouse strains.


Sujet(s)
Encéphale/effets des médicaments et des substances chimiques , Éthanol/pharmacologie , Récepteur dopamine D1/effets des médicaments et des substances chimiques , Récepteur D2 de la dopamine/effets des médicaments et des substances chimiques , Récompense , Autostimulation/effets des médicaments et des substances chimiques , Animaux , Benzazépines/pharmacologie , Encéphale/physiologie , Stimulation électrique , Mâle , Souris , Souris de lignée C57BL , Souris de lignée DBA , Quinpirole/pharmacologie , Raclopride/pharmacologie , Récepteur dopamine D1/physiologie , Récepteur D2 de la dopamine/physiologie
16.
Psychopharmacology (Berl) ; 231(17): 3415-3423, 2014 Sep.
Article de Anglais | MEDLINE | ID: mdl-24810108

RÉSUMÉ

RATIONALE: The neuroactive steroid (3α,5α)-3-hydroxy-pregnan-20-one (3α,5α-THP, allopregnanolone) has effects on reward-related behaviors in mice and rats that suggest that it may activate brain reward circuits. Intracranial self-stimulation (ICSS) is an operant behavioral technique that detects changes in the sensitivity of brain reward circuitry following drug administration. OBJECTIVE: To examine the effects of the neuroactive steroid allopregnanolone on ICSS and to compare these effects to those of cocaine. METHODS: Male C57BL/6J mice implanted with stimulating electrodes implanted into the medial forebrain bundle responded for reinforcement by electrical stimulation (brain stimulation reward (BSR)). Mice received cocaine (n = 11, 3.0-30.0 mg/kg, intraperitoneal (i.p.)) or the neuroactive steroid allopregnanolone (n = 11, 3.0-17.0 mg/kg, i.p.). BSR thresholds (θ 0) and maximum (MAX) operant response rates after drug treatments were compared to those after vehicle injections. RESULTS: Cocaine and allopregnanolone dose dependently lowered BSR thresholds relative to vehicle injections. Cocaine was maximally effective (80 % reduction) in the second 15 min following the 30 mg/kg dose, while allopregnanolone was maximally effective (30 % reduction) 15-45 min after the 17 mg/kg dose. Neither drug had significant effects on MAX response rates. CONCLUSIONS: The effects of allopregnanolone on BSR thresholds are consistent with the previously reported effects of benzodiazepines and alcohol, suggesting that positive modulation of GABAA receptors can facilitate reward-related behaviors in C57BL/6J mice.


Sujet(s)
Encéphale/effets des médicaments et des substances chimiques , Agents neuromédiateurs/pharmacologie , Prégnanolone/pharmacologie , Autostimulation/effets des médicaments et des substances chimiques , Animaux , Cocaïne/pharmacologie , Troubles liés à la cocaïne/psychologie , Relation dose-effet des médicaments , Mâle , Souris , Souris de lignée C57BL , Récompense
17.
Behav Pharmacol ; 25(1): 61-70, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24322822

RÉSUMÉ

The antiepileptic levetiracetam (LEV) has been investigated for the treatment of alcohol abuse. However, little is known about how LEV alters the behavioral effects of alcohol in laboratory animals. The acute effects of LEV on alcohol drinking by male C57BL/6J mice were investigated using two different drinking procedures, limited access [drinking-in-the-dark (DID)] and intermittent access (IA) drinking. In the first experiment (DID), mice had access to a single bottle containing alcohol or sucrose for 4 h every other day. In the second experiment (IA), mice had IA to two bottles, one containing alcohol or sucrose and one containing water, for 24 h on Monday, Wednesday, and Friday. In both experiments, mice were administered LEV (0.3-100 mg/kg intraperitoneally) or vehicle 30 min before access to the drinking solutions. In the DID mice, LEV increased alcohol intake from 4.3 to 5.4 g/kg, whereas in the IA mice LEV decreased alcohol intake from 4.8 to 3.0 g/kg in the first 4 h of access and decreased 24 h alcohol intake from 20 to ∼15 g/kg. These effects appear specific to alcohol, as LEV did not affect sucrose intake in either experiment. LEV appears to differentially affect drinking in animal models of moderate and heavier alcohol consumption.


Sujet(s)
Consommation d'alcool/traitement médicamenteux , Anticonvulsivants/pharmacologie , Comportement de choix/effets des médicaments et des substances chimiques , Piracétam/analogues et dérivés , Consommation d'alcool/psychologie , Animaux , Dépresseurs du système nerveux central/administration et posologie , Dépresseurs du système nerveux central/sang , Relation dose-effet des médicaments , Comportement dipsique/effets des médicaments et des substances chimiques , Comportement dipsique/physiologie , Éthanol/administration et posologie , Éthanol/sang , Lévétiracétam , Mâle , Souris , Souris de lignée C57BL , Piracétam/pharmacologie , Saccharose/administration et posologie , Édulcorants/administration et posologie
18.
PLoS One ; 8(10): e77896, 2013.
Article de Anglais | MEDLINE | ID: mdl-24205018

RÉSUMÉ

Fragile X syndrome (FXS) is a leading cause of intellectual disability. FXS is caused by loss of function of the FMR1 gene, and mice in which Fmr1 has been inactivated have been used extensively as a preclinical model for FXS. We investigated the behavioral pharmacology of drugs acting through dopaminergic, glutamatergic, and cholinergic systems in fragile X (Fmr1 (-/Y)) mice with intracranial self-stimulation (ICSS) and locomotor activity measurements. We also measured brain expression of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine biosynthesis. Fmr1 (-/Y) mice were more sensitive than wild type mice to the rewarding effects of cocaine, but less sensitive to its locomotor stimulating effects. Anhedonic but not motor depressant effects of the atypical neuroleptic, aripiprazole, were reduced in Fmr1 (-/Y) mice. The mGluR5-selective antagonist, 6-methyl-2-(phenylethynyl)pyridine (MPEP), was more rewarding and the preferential M1 antagonist, trihexyphenidyl, was less rewarding in Fmr1 (-/Y) than wild type mice. Motor stimulation by MPEP was unchanged, but stimulation by trihexyphenidyl was markedly increased, in Fmr1 (-/Y) mice. Numbers of midbrain TH+ neurons in the ventral tegmental area were unchanged, but were lower in the substantia nigra of Fmr1 (-/Y) mice, although no changes in TH levels were found in their forebrain targets. The data are discussed in the context of known changes in the synaptic physiology and pharmacology of limbic motor systems in the Fmr1 (-/Y) mouse model. Preclinical findings suggest that drugs acting through multiple neurotransmitter systems may be necessary to fully address abnormal behaviors in individuals with FXS.


Sujet(s)
Comportement animal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Inhibiteurs de la capture de la dopamine/pharmacologie , Antagonistes des acides aminés excitateurs/pharmacologie , Syndrome du chromosome X fragile/traitement médicamenteux , Antagonistes muscariniques/pharmacologie , Récompense , Animaux , Neuroleptiques/pharmacologie , Aripiprazole , Technique de Western , Cocaïne/pharmacologie , Protéine du syndrome X fragile/physiologie , Syndrome du chromosome X fragile/métabolisme , Techniques immunoenzymatiques , Mâle , Souris , Souris knockout , Activité motrice/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie , Pyridines/pharmacologie , Quinolinone/pharmacologie , Récepteur-5 métabotropique du glutamate/antagonistes et inhibiteurs , Récepteur-5 métabotropique du glutamate/métabolisme , Trihexyphénidyle/pharmacologie , Tyrosine 3-monooxygenase/métabolisme
19.
Neuropsychopharmacology ; 38(7): 1322-33, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23353709

RÉSUMÉ

The antiepileptic drug levetiracetam (LEV) is a potential treatment for alcohol use disorders, yet few preclinical studies exist on its effects in animal models relevant to drug or alcohol abuse. We investigated the effects of LEV on locomotor stimulation following acute and repeated administration of alcohol or cocaine and on alcohol- and cocaine-mediated changes in responding for brain stimulation reward (BSR) in C57BL/6J mice. LEV alone (10.0-100.0 mg/kg intraperitoneally) had no significant effect on locomotor activity or intracranial self-stimulation. Pretreatment with LEV reduced acute locomotor stimulation by 2.0 g/kg alcohol, attenuated the development of locomotor sensitization to alcohol with repeated exposure, and produced a shift in the dose-response curve for alcohol on BSR threshold without affecting maximum operant response rate (MAX). Conversely, LEV pretreatment enhanced both acute locomotor stimulation by 15 mg/kg cocaine and development of locomotor sensitization following repeated exposure and produced a leftward shift in the dose-response curve for cocaine on BSR threshold without affecting MAX. Electrophysiological recordings in vitro showed that LEV reduced excitatory currents in both ventral tegmental area (VTA) dopamine neurons and nucleus accumbens (NAc) medium spiny neurons, consistent with a presynaptic effect. The opposite effects of LEV pretreatment on alcohol- and cocaine-related behaviors may predict its clinical utility in the treatment of patients with alcohol, but not psychostimulant abuse disorders.


Sujet(s)
Cocaïne/agonistes , Éthanol/antagonistes et inhibiteurs , Activité motrice/effets des médicaments et des substances chimiques , Piracétam/analogues et dérivés , Autostimulation/effets des médicaments et des substances chimiques , Animaux , Anticonvulsivants/pharmacologie , Dépresseurs du système nerveux central/antagonistes et inhibiteurs , Dépresseurs du système nerveux central/pharmacologie , Sensibilisation du système nerveux central/effets des médicaments et des substances chimiques , Cocaïne/pharmacologie , Inhibiteurs de la capture de la dopamine/agonistes , Inhibiteurs de la capture de la dopamine/pharmacologie , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Neurones dopaminergiques/physiologie , Relation dose-effet des médicaments , Interactions médicamenteuses , Éthanol/pharmacologie , Potentiels post-synaptiques excitateurs/effets des médicaments et des substances chimiques , Lévétiracétam , Mâle , Souris , Neurones/effets des médicaments et des substances chimiques , Neurones/physiologie , Noyau accumbens/effets des médicaments et des substances chimiques , Noyau accumbens/physiologie , Piracétam/pharmacologie , Aire tegmentale ventrale/effets des médicaments et des substances chimiques , Aire tegmentale ventrale/physiologie
20.
J Clin Invest ; 122(12): 4544-54, 2012 Dec.
Article de Anglais | MEDLINE | ID: mdl-23143301

RÉSUMÉ

Angelman syndrome (AS) is a neurodevelopmental disorder caused by maternal deletions or mutations of the ubiquitin ligase E3A (UBE3A) allele and characterized by minimal verbal communication, seizures, and disorders of voluntary movement. Previous studies have suggested that abnormal dopamine neurotransmission may underlie some of these deficits, but no effective treatment currently exists for the core features of AS. A clinical trial of levodopa (L-DOPA) in AS is ongoing, although the underlying rationale for this treatment strategy has not yet been thoroughly examined in preclinical models. We found that AS model mice lacking maternal Ube3a (Ube3a(m-/p+) mice) exhibit behavioral deficits that correlated with abnormal dopamine signaling. These deficits were not due to loss of dopaminergic neurons or impaired dopamine synthesis. Unexpectedly, Ube3a(m-/p+) mice exhibited increased dopamine release in the mesolimbic pathway while also exhibiting a decrease in dopamine release in the nigrostriatal pathway, as measured with fast-scan cyclic voltammetry. These findings demonstrate the complex effects of UBE3A loss on dopamine signaling in subcortical motor pathways that may inform ongoing clinical trials of L-DOPA therapy in patients with AS.


Sujet(s)
Syndrome d'Angelman/métabolisme , Dopamine/physiologie , Neurones dopaminergiques/métabolisme , Transmission synaptique , Syndrome d'Angelman/anatomopathologie , Animaux , Benzazépines/pharmacologie , Cocaïne/pharmacologie , Modèles animaux de maladie humaine , Dopamine/métabolisme , Antagonistes du récepteur D2 de la dopamine , Inhibiteurs de la capture de la dopamine/pharmacologie , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Stimulation électrique , Femelle , Indoles/pharmacologie , Mâle , Souris , Souris de lignée C57BL , Activité motrice/effets des médicaments et des substances chimiques , Pipéridines/pharmacologie , Raclopride/pharmacologie , Récepteur dopamine D1/antagonistes et inhibiteurs , Récompense , Autostimulation , Substantia nigra/métabolisme , Substantia nigra/anatomopathologie , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Aire tegmentale ventrale/métabolisme , Aire tegmentale ventrale/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...