Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Radiat Res ; 188(5): 532-551, 2017 11.
Article de Anglais | MEDLINE | ID: mdl-28945526

RÉSUMÉ

Astronauts traveling to Mars will be exposed to chronic low doses of galactic cosmic space radiation, which contains highly charged, high-energy (HZE) particles. 56Fe-HZE-particle exposure decreases hippocampal dentate gyrus (DG) neurogenesis and disrupts hippocampal function in young adult rodents, raising the possibility of impaired astronaut cognition and risk of mission failure. However, far less is known about how exposure to other HZE particles, such as 28Si, influences hippocampal neurogenesis and function. To compare the influence of 28Si exposure on indices of neurogenesis and hippocampal function with previous studies on 56Fe exposure, 9-week-old C57BL/6J and Nestin-GFP mice (NGFP; made and maintained for 10 or more generations on a C57BL/6J background) received whole-body 28Si-particle-radiation exposure (0, 0.2 and 1 Gy, 300 MeV/n, LET 67 KeV/µ, dose rate 1 Gy/min). For neurogenesis assessment, the NGFP mice were injected with the mitotic marker BrdU at 22 h postirradiation and brains were examined for indices of hippocampal proliferation and neurogenesis, including Ki67+, BrdU+, BrdU+NeuN+ and DCX+ cell numbers at short- and long-term time points (24 h and 3 months postirradiation, respectively). In the short-term group, stereology revealed fewer Ki67+, BrdU+ and DCX+ cells in 1-Gy-irradiated group relative to nonirradiated control mice, fewer Ki67+ and DCX+ cells in 0.2 Gy group relative to control group and fewer BrdU+ and DCX+ cells in 1 Gy group relative to 0.2 Gy group. In contrast to the clearly observed radiation-induced, dose-dependent reductions in the short-term group across all markers, only a few neurogenesis indices were changed in the long-term irradiated groups. Notably, there were fewer surviving BrdU+ cells in the 1 Gy group relative to 0- and 0.2-Gy-irradiated mice in the long-term group. When the short- and long-term groups were analyzed by sex, exposure to radiation had a similar effect on neurogenesis indices in male and female mice, although only male mice showed fewer surviving BrdU+ cells in the long-term group. Fluorescent immunolabeling and confocal phenotypic analysis revealed that most surviving BrdU+ cells in the long-term group expressed the neuronal marker NeuN, definitively confirming that exposure to 1 Gy 28Si radiation decreased the number of surviving adult-generated neurons in male mice relative to both 0- and 0.2-Gy-irradiated mice. For hippocampal function assessment, 9-week-old male C57BL/6J mice received whole-body 28Si-particle exposure and were then assessed long-term for performance on contextual and cued fear conditioning. In the context test the animals that received 0.2 Gy froze less relative to control animals, suggesting decreased hippocampal-dependent function. However, in the cued fear conditioning test, animals that received 1 Gy froze more during the pretone portion of the test, relative to controls and 0.2-Gy-irradiated mice, suggesting enhanced anxiety. Compared to previously reported studies, these data suggest that 28Si-radiation exposure damages neurogenesis, but to a lesser extent than 56Fe radiation and that low-dose 28Si exposure induces abnormalities in hippocampal function, disrupting fear memory but also inducing anxiety-like behavior. Furthermore, exposure to 28Si radiation decreased new neuron survival in long-term male groups but not females suggests that sex may be an important factor when performing brain health risk assessment for astronauts traveling in space.


Sujet(s)
Conditionnement psychologique/effets des radiations , Gyrus denté/cytologie , Peur/psychologie , Neurogenèse/effets des radiations , Neurones/cytologie , Silicium , Irradiation corporelle totale/effets indésirables , Animaux , Comportement animal/physiologie , Comportement animal/effets des radiations , Prolifération cellulaire/effets des radiations , Survie cellulaire/effets des radiations , Rayonnement cosmique , Gyrus denté/physiologie , Gyrus denté/effets des radiations , Relation dose-effet des rayonnements , Protéine doublecortine , Peur/effets des radiations , Femelle , Mémoire/physiologie , Mémoire/effets des radiations , Souris , Neurones/effets des radiations , Facteurs temps
2.
Article de Anglais | MEDLINE | ID: mdl-26627453

RÉSUMÉ

Aging is strongly correlated with decreases in neurogenesis, the process by which neural stem and progenitor cells proliferate and differentiate into new neurons. In addition to stem-cell-intrinsic factors that change within the aging stem-cell pool, recent evidence emphasizes new roles for systemic and microenvironmental factors in modulating the neurogenic niche. This article focuses on new insights gained through the use of heterochronic parabiosis models, in which an old mouse and a young circulatory system are joined. By studying the brains of both young and old mice, researchers are beginning to uncover circulating proneurogenic "youthful" factors and "aging" factors that decrease stem-cell activity and neurogenesis. Ultimately, the identification of factors that influence stem-cell aging may lead to strategies that slow or even reverse age-related decreases in neural-stem-cell (NSC) function and neurogenesis.


Sujet(s)
Vieillissement/physiologie , Prolifération cellulaire/physiologie , Cellules souches neurales/physiologie , Niche de cellules souches/physiologie , Animaux , Encéphale/physiologie , Humains , Souris , Neurogenèse , Neurones/physiologie , Parabiose
3.
Life Sci Space Res (Amst) ; 2: 70-79, 2014 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-25170435

RÉSUMÉ

The high-LET HZE particles from galactic cosmic radiation pose tremendous health risks to astronauts, as they may incur sub-threshold brain injury or maladaptations that may lead to cognitive impairment. The health effects of HZE particles are difficult to predict and unfeasible to prevent. This underscores the importance of estimating radiation risks to the central nervous system as a whole as well as to specific brain regions like the hippocampus, which is central to learning and memory. Given that neurogenesis in the hippocampus has been linked to learning and memory, we investigated the response and recovery of neurogenesis and neural stem cells in the adult mouse hippocampal dentate gyrus after HZE particle exposure using two nestin transgenic reporter mouse lines to label and track radial glia stem cells (Nestin-GFP and Nestin-CreERT2/R26R:YFP mice, respectively). Mice were subjected to 56Fe particle exposure (0 or 1 Gy, at either 300 or 1000 MeV/n) and brains were harvested at early (24h), intermediate (7d), and/or long time points (2-3mo) post-irradiation. 56Fe particle exposure resulted in a robust increase in 53BP1+ foci at both the intermediate and long time points post-irradiation, suggesting long-term genomic instability in the brain. However, 56Fe particle exposure only produced a transient decrease in immature neuron number at the intermediate time point, with no significant decrease at the long time point post-irradiation. 56Fe particle exposure similarly produced a transient decrease in dividing progenitors, with fewer progenitors labeled at the early time point but equal number labeled at the intermediate time point, suggesting a recovery of neurogenesis. Notably, 56Fe particle exposure did not change the total number of nestin-expressing neural stem cells. These results highlight that despite the persistence of an index of genomic instability, 56Fe particle-induced deficits in adult hippocampal neurogenesis may be transient. These data support the regenerative capacity of the adult SGZ after HZE particle exposure and encourage additional inquiry into the relationship between radial glia stem cells and cognitive function after HZE particle exposure.

4.
Radiat Res ; 180(6): 658-67, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-24320054

RÉSUMÉ

Astronauts on multi-year interplanetary missions will be exposed to a low, chronic dose of high-energy, high-charge particles. Studies in rodents show acute, nonfractionated exposure to these particles causes brain changes such as fewer adult-generated hippocampal neurons and stem cells that may be detrimental to cognition and mood regulation and thus compromise mission success. However, the influence of a low, chronic dose of these particles on neurogenesis and stem cells is unknown. To examine the influence of galactic cosmic radiation on neurogenesis, adult-generated stem and progenitor cells in Nestin-CreER(T2)/R26R-YFP transgenic mice were inducibly labeled to allow fate tracking. Mice were then sham exposed or given one acute 100 cGy (56)Fe-particle exposure or five fractionated 20 cGy (56)Fe-particle exposures. Adult-generated hippocampal neurons and stem cells were quantified 24 h or 3 months later. Both acute and fractionated exposure decreased the amount of proliferating cells and immature neurons relative to sham exposure. Unexpectedly, neither acute nor fractionated exposure decreased the number of adult neural stem cells relative to sham expsoure. Our findings show that single and fractionated exposures of (56)Fe-particle irradiation are similarly detrimental to adult-generated neurons. Implications for future missions and ground-based studies in space radiation are discussed.


Sujet(s)
Hippocampe/cytologie , Hippocampe/effets des radiations , Fer , Transfert linéique d'énergie , Neurogenèse/effets des radiations , Animaux , Numération cellulaire , Prolifération cellulaire/effets des radiations , Gyrus denté/cytologie , Gyrus denté/effets des radiations , Fractionnement de la dose d'irradiation , Femelle , Mâle , Souris , Neurones/cytologie , Neurones/effets des radiations , Appréciation des risques , Facteurs temps
5.
Hippocampus ; 23(8): 708-19, 2013 Aug.
Article de Anglais | MEDLINE | ID: mdl-23554226

RÉSUMÉ

Radial glia-like cells (RGCs) are the hypothesized source of adult hippocampal neurogenesis. However, the current model of hippocampal neurogenesis does not fully incorporate the in vivo heterogeneity of RGCs. In order to better understand the contribution of different RGC subtypes to adult hippocampal neurogenesis, we employed widely used transgenic lines (Nestin-CreER(T2) and GLAST::CreER(T2) mice) to explore how RGCs contribute to neurogenesis under basal conditions and after stimulation and depletion of neural progenitor cells. We first used these inducible fate-tracking transgenic lines to define the similarities and differences in the contribution of nestin- and GLAST-lineage cells to basal long-term hippocampal neurogenesis. We then explored the ability of nestin- and GLAST-lineage RGCs to contribute to neurogenesis after experimental manipulations that either ablate neurogenesis (i.c.v. application of the anti-mitotic AraC, cytosine-ß-D-arabinofuranoside) or stimulate neurogenesis (wheel running). Interestingly, in both ablation and stimulation experiments, labeled RGCs in GLAST::CreER(T2) mice appear to contribute to neurogenesis, whereas RGCs in Nestin-CreER(T2) mice do not. Finally, using NestinGFP reporter mice, we expanded on previous research by showing that not all RGCs in the adult dentate gyrus subgranular zone express nestin, and therefore RGCs are antigenically heterogeneous. These findings are important for the field, as they allow appropriately conservative interpretation of existing and future data that emerge from these inducible transgenic lines. These findings also raise important questions about the differences between transgenic driver lines, the heterogeneity of RGCs, and the potential differences in progenitor cell behavior between transgenic lines. As these findings highlight the possible differences in the contribution of cells to long-term neurogenesis in vivo, they indicate that the current models of hippocampal neurogenesis should be modified to include RGC lineage heterogeneity.


Sujet(s)
Lignage cellulaire/physiologie , Transporteur-1 d'acides aminés excitateurs/métabolisme , Hippocampe/cytologie , Nestine/métabolisme , Neurogenèse/physiologie , Animaux , Protéines à domaine doublecortine , Transporteur-1 d'acides aminés excitateurs/génétique , Protéine-7 de liaison aux acides gras , Protéines de liaison aux acides gras/métabolisme , Protéine gliofibrillaire acide/métabolisme , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Souris , Souris de lignée C57BL , Souris transgéniques , Protéines associées aux microtubules/métabolisme , Activité motrice/physiologie , Protéines de tissu nerveux/métabolisme , Nestine/génétique , Névroglie/physiologie , Neurones/physiologie , Neuropeptides/métabolisme , Techniques de culture d'organes , Facteurs de transcription SOX-B1/métabolisme , Cellules souches/physiologie
6.
J Neurosci ; 30(31): 10484-92, 2010 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-20685991

RÉSUMÉ

Notch1 regulates neural stem cell (NSC) number during development, but its role in adult neurogenesis is unclear. We generated nestin-CreER(T2)/R26R-YFP/Notch1(loxP/loxP) [Notch1inducible knock-out (iKO)] mice to allow tamoxifen (TAM)-inducible elimination of Notch1 and concomitant expression of yellow fluorescent protein (YFP) in nestin-expressing Type-1 NSCs and their progeny in the adult hippocampal subgranular zone (SGZ). Consistent with previous research, YFP+ cells in all stages of neurogenesis were evident in the subgranular zone (SGZ) of wild-type (WT) mice (nestin-CreER(T2)/R26R-YFP/Notch1(w/w)) after tamoxifen (post-TAM), producing adult-generated YFP+ dentate gyrus neurons. Compared with WT littermates, Notch1 iKO mice had similar numbers of total SGZ YFP+ cells 13 and 30 d post-TAM but had significantly fewer SGZ YFP+ cells 60 and 90 d post-TAM. Significantly fewer YFP+ Type-1 NSCs and transiently amplifying progenitors (TAPs) resulted in generation of fewer YFP+ granule neurons in Notch1 iKO mice. Strikingly, 30 d of running rescued this deficit, as the total YFP+ cell number in Notch iKO mice was equivalent to WT levels. This was even more notable given the persistent deficits in the Type-1 NSC and TAP reservoirs. Our data show that Notch1 signaling is required to maintain a reservoir of undifferentiated cells and ensure continuity of adult hippocampal neurogenesis, but that alternative Notch- and Type-1 NSC-independent pathways compensate in response to physical activity. These data shed light on the complex relationship between Type-1 NSCs, adult neurogenesis, the neurogenic niche, and environmental stimuli.


Sujet(s)
Cellules souches adultes/métabolisme , Hippocampe/métabolisme , Neurogenèse/physiologie , Récepteur Notch1/métabolisme , Animaux , Numération cellulaire , Immunohistochimie , Souris , Souris transgéniques , Neurones/métabolisme , Conditionnement physique d'animal/physiologie
7.
Proc Natl Acad Sci U S A ; 107(9): 4436-41, 2010 Mar 02.
Article de Anglais | MEDLINE | ID: mdl-20176946

RÉSUMÉ

The long-term response to chronic stress is variable, with some individuals developing maladaptive functioning, although other "resilient" individuals do not. Stress reduces neurogenesis in the dentate gyrus subgranular zone (SGZ), but it is unknown if stress-induced changes in neurogenesis contribute to individual vulnerability. Using a chronic social defeat stress model, we explored whether the susceptibility to stress-induced social avoidance was related to changes in SGZ proliferation and neurogenesis. Immediately after social defeat, stress-exposed mice (irrespective of whether they displayed social avoidance) had fewer proliferating SGZ cells labeled with the S-phase marker BrdU. The decrease was transient, because BrdU cell numbers were normalized 24 h later. The survival of BrdU cells labeled before defeat stress was also not altered. However, 4 weeks later, mice that displayed social avoidance had more surviving dentate gyrus neurons. Thus, dentate gyrus neurogenesis is increased after social defeat stress selectively in mice that display persistent social avoidance. Supporting a functional role for adult-generated dentate gyrus neurons, ablation of neurogenesis via cranial ray irradiation robustly inhibited social avoidance. These data show that the time window after cessation of stress is a critical period for the establishment of persistent cellular and behavioral responses to stress and that a compensatory enhancement in neurogenesis is related to the long-term individual differences in maladaptive responses to stress.


Sujet(s)
Apprentissage par évitement , Hippocampe/anatomopathologie , Neurogenèse , Stress psychologique/anatomopathologie , Animaux , Facteur neurotrophique dérivé du cerveau/métabolisme , Broxuridine/métabolisme , Mort cellulaire , Hippocampe/métabolisme , Mâle , Souris , Phase S , Transduction du signal
8.
Neuropharmacology ; 58(6): 884-93, 2010 May.
Article de Anglais | MEDLINE | ID: mdl-20060007

RÉSUMÉ

Over one-quarter of adult Americans are diagnosed with a mental illness like Major Depressive Disorder (MDD), Post-Traumatic Stress Disorder (PTSD), schizophrenia, and Alzheimer's Disease. In addition to the exceptional personal burden these disorders exert on patients and their families, they also have enormous cost to society. Although existing pharmacological and psychosocial treatments alleviate symptoms in many patients, the comorbidity, severity, and intractable nature of mental disorders strongly underscore the need for novel strategies. As the hippocampus is a site of structural and functional pathology in most mental illnesses, a hippocampal-based treatment approach has been proposed to counteract the cognitive deficits and mood dysregulation that are hallmarks of psychiatric disorders. In particular, preclinical and clinical research suggests that hippocampal neurogenesis, the generation of new neurons in the adult dentate gyrus, may be harnessed to treat mental illness. There are obvious applications and allures of this approach; for example, perhaps stimulating hippocampal neurogenesis would reverse the overt and noncontroversial hippocampal atrophy and functional deficits observed in Alzheimer's Disease and schizophrenia, or the more controversial hippocampal deficits seen in MDD and PTSD. However, critical examination suggests that neurogenesis may only correlate with mental illness and treatment, suggesting targeting neurogenesis alone is not a sufficient treatment strategy. Here we review the classic and causative links between adult hippocampal neurogenesis and mental disorders, and provide a critical evaluation of how (and if) our basic knowledge of new neurons in the adult hippocampus might eventually help combat or even prevent mental illness.


Sujet(s)
Hippocampe/physiopathologie , Troubles mentaux/physiopathologie , Troubles mentaux/thérapie , Neurogenèse/physiologie , Cellules souches adultes/physiologie , Animaux , Humains , Neurones/physiologie
9.
Bioessays ; 30(2): 102-6, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-18200563

RÉSUMÉ

Critical cellular functions, including stem cell maintenance, fate determination, and cellular behavior, are governed by canonical Wnt signaling, an evolutionarily conserved pathway whose intracellular signal is transduced by beta-catentin. Emerging evidence suggests that canonical Wnt signaling influences cellular aging, indicating that increases in Wnt signaling delay age-related deficits.1 However, recent Science papers suggest that Wnt signaling accelerates the onset of aging.2,3 In an attempt to resolve this paradox and clarify how Wnt signaling affects aging, we provide a selective review of research relevant to Wnt signaling and aging.


Sujet(s)
Vieillissement de la cellule/physiologie , Transduction du signal/physiologie , Protéines de type Wingless/physiologie , Animaux , Humains , Modèles biologiques , Protéines de type Wingless/métabolisme
10.
J Neurosci ; 27(46): 12623-9, 2007 Nov 14.
Article de Anglais | MEDLINE | ID: mdl-18003841

RÉSUMÉ

Understanding the fate of adult-generated neurons and the mechanisms that influence them requires consistent labeling and tracking of large numbers of stem cells. We generated a nestin-CreER(T2)/R26R-yellow fluorescent protein (YFP) mouse to inducibly label nestin-expressing stem cells and their progeny in the adult subventricular zone (SVZ) and subgranular zone (SGZ). Several findings show that the estrogen ligand tamoxifen (TAM) specifically induced recombination in stem cells and their progeny in nestin-CreER(T2)/R26R-YFP mice: 97% of SGZ stem-like cells (GFAP/Sox2 with radial glial morphology) expressed YFP; YFP+ neurospheres could be generated in vitro after recombination in vivo, and maturing YFP+ progeny were increasingly evident in the olfactory bulb (OB) and dentate gyrus (DG) granule cell layer. Revealing an unexpected regional dissimilarity in adult neurogenesis, YFP+ cells accumulated up to 100 d after TAM in the OB, but in the SGZ, YFP+ cells reached a plateau 30 d after TAM. In addition, most SVZ and SGZ YFP+ cells became neurons, underscoring a link between nestin and neuronal fate. Finally, quantification of YFP+ cells in nestin-CreER(T2)/R26R-YFP mice allowed us to estimate, for example, that stem cells and their progeny contribute to no more than 1% of the adult DG granule cell layer. In addition to revealing the dynamic contribution of nestin-expressing stem cells to adult neurogenesis, this work highlights the utility of the nestin-CreER(T2)/R26R-YFP mouse for inducible gene ablation in stem cells and their progeny in vivo in the two major regions of adult neurogenesis.


Sujet(s)
Encéphale/métabolisme , Différenciation cellulaire/physiologie , Lignage cellulaire/physiologie , Protéines de filaments intermédiaires/métabolisme , Protéines de tissu nerveux/métabolisme , Neurones/métabolisme , Cellules souches/métabolisme , Animaux , Encéphale/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignage cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Gyrus denté/cytologie , Gyrus denté/métabolisme , Ciblage de gène/méthodes , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Souris , Souris transgéniques , Modèles animaux , Régénération nerveuse/effets des médicaments et des substances chimiques , Régénération nerveuse/physiologie , Nestine , Névroglie/cytologie , Névroglie/effets des médicaments et des substances chimiques , Névroglie/métabolisme , Bulbe olfactif/cytologie , Bulbe olfactif/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Recombinaison génétique/effets des médicaments et des substances chimiques , Recombinaison génétique/génétique , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie , Cellules souches/effets des médicaments et des substances chimiques , Tamoxifène/pharmacologie
11.
Nat Neurosci ; 10(1): 93-9, 2007 Jan.
Article de Anglais | MEDLINE | ID: mdl-17143271

RÉSUMÉ

Chronic morphine administration (via subcutaneous pellet) decreases the size of dopamine neurons in the ventral tegmental area (VTA), a key reward region in the brain, yet the molecular basis and functional consequences of this effect are unknown. In this study, we used viral-mediated gene transfer in rat to show that chronic morphine-induced downregulation of the insulin receptor substrate 2 (IRS2)-thymoma viral proto-oncogene (Akt) signaling pathway in the VTA mediates the decrease in dopamine cell size seen after morphine exposure and that this downregulation diminishes morphine reward, as measured by conditioned place preference. We further show that the reduction in size of VTA dopamine neurons persists up to 2 weeks after morphine withdrawal, which parallels the tolerance to morphine's rewarding effects caused by previous chronic morphine exposure. These findings directly implicate the IRS2-Akt signaling pathway as a critical regulator of dopamine cell morphology and opiate reward.


Sujet(s)
Conditionnement opérant/effets des médicaments et des substances chimiques , Dopamine/métabolisme , Mésencéphale/cytologie , Morphine/administration et posologie , Stupéfiants/administration et posologie , Neurones/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Technique de Western , Taille de la cellule/effets des médicaments et des substances chimiques , Conditionnement opérant/physiologie , Relation dose-effet des médicaments , Calendrier d'administration des médicaments , Antienzymes/pharmacologie , Immunohistochimie , Substrats du récepteur à l'insuline , Protéines et peptides de signalisation intracellulaire/physiologie , Mâle , Activité motrice/effets des médicaments et des substances chimiques , Activité motrice/physiologie , Neurones/physiologie , Protéine oncogène v-akt/physiologie , Phosphoprotéines/physiologie , Rats , Rat Sprague-Dawley , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs temps
12.
Pharmacol Biochem Behav ; 74(3): 755-63, 2003 Feb.
Article de Anglais | MEDLINE | ID: mdl-12543242

RÉSUMÉ

Strain-dependent differences have been used to highlight unknown genetic contributions to important behavioral and physiological end points. In this regard, the Fischer (F344) and Lewis (LEW) rat strains have often been studied because they exhibit a myriad of behavioral and physiological differences. Recently, schedule-induced polydipsia (SIP), a potential model of stress and drug abuse, has been reported to differ between the two strains (see [Pharmacol. Biochem. Behav. 67 (2002) 809]) with F344 rats displaying greater levels of consumption than LEW rats. Given the importance of SIP as a behavioral model of stress and of drug abuse, the present study further explored SIP in F344 and LEW strains by assessing the acquisition and steady-state performance of SIP (under a fixed-time 30 schedule of food delivery; FT30), its characteristic postprandial temporal licking pattern and its modulation by variations in the food delivery schedule (FT15, FT30 and FT60). F344 rats acquired SIP at a faster rate and drank at a higher asymptotic level than LEW rats. Both strains displayed the typical inverted U-shaped post-pellet pattern of drinking and changes in levels of consumption (and displacement of the initiation of post-pellet drinking) with changes in the FT value, supporting the position that the increased drinking seen in both groups was schedule induced. These strain differences in SIP are consistent with the fact that the F344 and LEW strains differ on other behavioral and physiological indices of stress and raise the issue of the use of this model in the assessment of differential drug intake between the two strains.


Sujet(s)
Consommation de boisson/physiologie , Programme de renforcement , Stress physiologique/génétique , Stress physiologique/psychologie , Soif/physiologie , Animaux , Consommation de boisson/génétique , Consommation alimentaire/génétique , Femelle , Rats , Rats de lignée F344 , Rats de lignée LEW , Spécificité d'espèce
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...