Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.806
Filtrar
1.
Alcohol Alcohol ; 59(4)2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38742547

RESUMEN

AIMS: Continued alcohol consumption despite negative consequences is a core symptom of alcohol use disorder. This is modeled in mice by pairing negative stimuli with alcohol, such as adulterating alcohol solution with quinine. Mice consuming alcohol under these conditions are considered to be engaging in aversion-resistant intake. Previously, we have observed sex differences in this behavior, with females more readily expressing aversion-resistant consumption. We also identified three brain regions that exhibited sex differences in neuronal activation during quinine-alcohol drinking: ventromedial prefrontal cortex (vmPFC), posterior insular cortex (PIC), and ventral tegmental area (VTA). Specifically, male mice showed increased activation in vmPFC and PIC, while females exhibited increased activation in VTA. In this study, we aimed to identify what specific type of neurons are activated in these regions during quinine-alcohol drinking. METHOD: We assessed quinine-adulterated alcohol intake using the two-bottle choice procedure. We also utilized RNAscope in situ hybridization in the three brain regions that previously exhibited a sex difference to examine colocalization of Fos, glutamate, GABA, and dopamine. RESULT: Females showed increased aversion-resistant alcohol consumption compared to males. We also found that males had higher colocalization of glutamate and Fos in vmPFC and PIC, while females had greater dopamine and Fos colocalization in the VTA. CONCLUSIONS: Collectively, these experiments suggest that glutamatergic output from the vmPFC and PIC may have a role in suppressing, and dopaminergic activity in the VTA may promote, aversion-resistant alcohol consumption. Future experiments will examine neuronal circuits that contribute to sex differences in aversion resistant consumption.


Asunto(s)
Consumo de Bebidas Alcohólicas , Neuronas , Quinina , Caracteres Sexuales , Animales , Quinina/farmacología , Femenino , Masculino , Ratones , Neuronas/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , Mesencéfalo/metabolismo , Mesencéfalo/efectos de los fármacos , Corteza Insular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Etanol/farmacología , Ácido Glutámico/metabolismo
2.
Addict Biol ; 29(5): e13397, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38711205

RESUMEN

Neuronal ensembles in the medial prefrontal cortex mediate cocaine self-administration via projections to the nucleus accumbens. We have recently shown that neuronal ensembles in the prelimbic cortex form rapidly to mediate cocaine self-administration. However, the role of neuronal ensembles within the nucleus accumbens in initial cocaine-seeking behaviour remains unknown. Here, we sought to expand the current literature by testing the necessity of the cocaine self-administration ensemble in the nucleus accumbens core (NAcCore) 1 day after male and female rats acquire cocaine self-administration by using the Daun02 inactivation procedure. We found that disrupting the NAcCore ensembles after a no-cocaine reward-seeking test increased subsequent cocaine seeking, while disrupting NAcCore ensembles following a cocaine self-administration session decreased subsequent cocaine seeking. We then characterized neuronal cell type in the NAcCore using RNAscope in situ hybridization. In the no-cocaine session, we saw reduced dopamine D1 type neuronal activation, while in the cocaine self-administration session, we found preferential dopamine D1 type neuronal activity in the NAcCore.


Asunto(s)
Cocaína , Comportamiento de Búsqueda de Drogas , Neuronas , Núcleo Accumbens , Autoadministración , Animales , Núcleo Accumbens/efectos de los fármacos , Cocaína/farmacología , Masculino , Femenino , Ratas , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Neuronas/efectos de los fármacos , Recompensa , Inhibidores de Captación de Dopamina/farmacología , Refuerzo en Psicología , Receptores de Dopamina D1 , Trastornos Relacionados con Cocaína/fisiopatología , Ratas Sprague-Dawley , Corteza Prefrontal/efectos de los fármacos
3.
Behav Pharmacol ; 35(4): 211-226, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38651984

RESUMEN

Stimulation of the innate immune system prior to stress exposure is a possible strategy to prevent depression under stressful conditions. Based on the innate immune system stimulating activities of zymosan A, we hypothesize that zymosan A may prevent the development of chronic stress-induced depression-like behavior. Our results showed that a single injection of zymosan A 1 day before stress exposure at a dose of 2 or 4 mg/kg, but not at a dose of 1 mg/kg, prevented the development of depression-like behaviors in mice treated with chronic social defeat stress (CSDS). The prophylactic effect of a single zymosan A injection (2 mg/kg) on CSDS-induced depression-like behaviors disappeared when the time interval between zymosan A and stress exposure was extended from 1 day or 5 days to 10 days, which was rescued by a second zymosan A injection 10 days after the first zymosan A injection and 4 days (4×, once daily) of zymosan A injections 10 days before stress exposure. Further analysis showed that a single zymosan A injection (2 mg/kg) 1 day before stress exposure could prevent the CSDS-induced increase in pro-inflammatory cytokines in the hippocampus and prefrontal cortex. Inhibition of the innate immune system by pretreatment with minocycline (40 mg/kg) abolished the preventive effect of zymosan A on CSDS-induced depression-like behaviors and CSDS-induced increase in pro-inflammatory cytokines in the brain. These results suggest that activation of the innate immune system triggered by zymosan A prevents the depression-like behaviors and neuroinflammatory responses in the brain induced by chronic stress.


Asunto(s)
Depresión , Hipocampo , Estrés Psicológico , Zimosan , Animales , Zimosan/farmacología , Ratones , Estrés Psicológico/inmunología , Masculino , Depresión/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Citocinas/metabolismo , Conducta Animal/efectos de los fármacos , Derrota Social , Inmunización/métodos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/inmunología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Minociclina/farmacología , Relación Dosis-Respuesta a Droga
4.
Environ Int ; 186: 108643, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38615544

RESUMEN

Exposure to bisphenol S (BPS) is known to adversely affect neuronal development. As pivotal components of neuronal polarization, axons and dendrites are indispensable structures within neurons, crucial for the maintenance of nervous system function. Here, we investigated the impact of BPS exposure on axonal and dendritic development both in vivo and in vitro. Our results revealed that exposure to BPS during pregnancy and lactation led to a reduction in the complexity, density, and length of axons and dendrites in the prefrontal cortex (PFC) of offspring. Employing RNA sequencing technology to elucidate the underlying mechanisms of axonal and dendritic damage induced by BPS, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis highlighted a significant alteration in the oxidative phosphorylation (OXPHOS) pathway, essential for mitochondrial function. Subsequent experiments demonstrate BPS-induced impairment in mitochondrial function, including damaged morphology, decreased adenosine triphosphate (ATP) and superoxide dismutase (SOD) levels, and increased reactive oxygen species and malondialdehyde (MDA). These alterations coincided with the downregulated expression of OXPHOS pathway-related genes (ATP6V1B1, ATP5K, NDUFC1, NDUFC2, NDUFA3, COX6B1) and Myosin 19 (Myo19). Notably, Myo19 overexpression restored the BPS-induced mitochondrial dysfunction by alleviating the inhibition of OXPHOS pathway. Consequently, this amelioration was associated with a reduction in BPS-induced axonal and dendritic injury observed in cultured neurons of the PFC.


Asunto(s)
Axones , Dendritas , Mitocondrias , Fosforilación Oxidativa , Fenoles , Sulfonas , Animales , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fenoles/toxicidad , Dendritas/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Femenino , Sulfonas/toxicidad , Axones/efectos de los fármacos , Embarazo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratones
5.
Eur J Pharmacol ; 972: 176559, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38588768

RESUMEN

This study aimed to assess whether brief recall of methamphetamine (MA) memory, when combined with ketamine (KE) treatment, may prevent stress-primed MA memory reinstatement. Combining 3-min recall and KE facilitated MA memory extinction and resistance to subsequent stress-primed reinstatement. Such combination also produced glutamate metabotropic receptor 5 (mGluR5) upregulation in animals' medial prefrontal cortex (mPFC) γ-amino-butyric acid (GABA) neuron. Accordingly, chemogenetic methods were employed to bi-directionally modulate mPFC GABA activity. Following brief recall and KE-produced MA memory extinction, intra-mPFC mDlx-Gi-coupled-human-muscarinic-receptor 4 (hM4Di)-infused mice receiving compound 21 (C21) treatment showed eminent stress-primed reinstatement, while their GABA mGluR5 expression seemed to be unaltered. Intra-mPFC mDlx-Gq-coupled-human-muscarinic-receptor 3 (hM3Dq)-infused mice undergoing C21 treatment displayed MA memory extinction and resistance to stress-provoked reinstatement. These results suggest that combining a brief recall and KE treatment and exciting mPFC GABA neuron may facilitate MA memory extinction and resistance to stress-primed recall. mPFC GABA neuronal activity plays a role in mediating brief recall/KE-produced effects on curbing the stress-provoked MA seeking.


Asunto(s)
Extinción Psicológica , Ketamina , Recuerdo Mental , Metanfetamina , Corteza Prefrontal , Receptor del Glutamato Metabotropico 5 , Estrés Psicológico , Animales , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Metanfetamina/farmacología , Ketamina/farmacología , Masculino , Ratones , Recuerdo Mental/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/psicología , Receptor del Glutamato Metabotropico 5/metabolismo , Extinción Psicológica/efectos de los fármacos , Memoria/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Ratones Endogámicos C57BL
6.
Behav Pharmacol ; 35(4): 193-200, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38567425

RESUMEN

Prepulse inhibition (PPI) is a crucial indicator of sensorimotor gating that is often impaired in neuropsychiatric diseases. Although dopamine D1 receptor agonists have been found to disrupt PPI in mice, the underlying mechanisms are not fully understood. In this study, we aimed to identify the brain regions responsible for the PPI-disruptive effect of the D1 agonist in mice. Results demonstrated that intraperitoneal administration of the selective dopamine D1 receptor agonist SKF82958 dramatically inhibited PPI, while the dopamine D1 receptor antagonist SCH23390 enhanced PPI. Additionally, local infusion of SKF82958 into the nucleus accumbens and medial prefrontal cortex disrupted PPI, but not in the ventral hippocampus. Infusion of SCH23390 into these brain regions also failed to enhance PPI. Overall, the study suggests that the nucleus accumbens and medial prefrontal cortex are responsible for the PPI-disruptive effect of dopamine D1 receptor agonists. These findings provide essential insights into the cellular and neural circuit mechanisms underlying the disruptive effects of dopamine D1 receptor agonists on PPI and may contribute to the development of novel treatments for neuropsychiatric diseases.


Asunto(s)
Benzazepinas , Agonistas de Dopamina , Ratones Endogámicos C57BL , Núcleo Accumbens , Corteza Prefrontal , Inhibición Prepulso , Receptores de Dopamina D1 , Animales , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Agonistas de Dopamina/farmacología , Ratones , Benzazepinas/farmacología , Masculino , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/metabolismo , Inhibición Prepulso/efectos de los fármacos , Inhibición Prepulso/fisiología , Reflejo de Sobresalto/efectos de los fármacos , Filtrado Sensorial/efectos de los fármacos , Antagonistas de Dopamina/farmacología
7.
Int J Mol Sci ; 25(8)2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38673763

RESUMEN

Chronic ethanol exposure often triggers neuroinflammation in the brain's reward system, potentially promoting the drive for ethanol consumption. A main marker of neuroinflammation is the microglia-derived monocyte chemoattractant protein 1 (MCP1) in animal models of alcohol use disorder in which ethanol is forcefully given. However, there are conflicting findings on whether MCP1 is elevated when ethanol is taken voluntarily, which challenges its key role in promoting motivation for ethanol consumption. Here, we studied MCP1 mRNA levels in areas implicated in consumption motivation-specifically, the prefrontal cortex, hippocampus, and striatum-as well as in the cerebellum, a brain area highly sensitive to ethanol, of C57BL/6 mice subjected to intermittent and voluntary ethanol consumption for two months. We found a significant increase in MCP1 mRNA levels in the cerebellum of mice that consumed ethanol compared to controls, whereas no significant changes were observed in the prefrontal cortex, hippocampus, or striatum or in microglia isolated from the hippocampus and striatum. To further characterize cerebellar neuroinflammation, we measured the expression changes in other proinflammatory markers and chemokines, revealing a significant increase in the proinflammatory microRNA miR-155. Notably, other classical proinflammatory markers, such as TNFα, IL6, and IL-1ß, remained unaltered, suggesting mild neuroinflammation. These results suggest that the onset of neuroinflammation in motivation-related areas is not required for high voluntary consumption in C57BL/6 mice. In addition, cerebellar susceptibility to neuroinflammation may be a trigger to the cerebellar degeneration that occurs after chronic ethanol consumption in humans.


Asunto(s)
Consumo de Bebidas Alcohólicas , Cerebelo , Quimiocina CCL2 , Cuerpo Estriado , Etanol , Hipocampo , Ratones Endogámicos C57BL , Corteza Prefrontal , Animales , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , Ratones , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Cerebelo/metabolismo , Cerebelo/efectos de los fármacos , Cerebelo/patología , Masculino , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Cuerpo Estriado/efectos de los fármacos , Etanol/efectos adversos , Consumo de Bebidas Alcohólicas/efectos adversos , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/patología , Microglía/metabolismo , Microglía/efectos de los fármacos , Microglía/patología , Inflamación/metabolismo , Inflamación/patología , Inflamación/inducido químicamente
8.
Brain Res Bull ; 211: 110935, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38570076

RESUMEN

Chronic ethanol consumption can lead to increased extracellular glutamate concentrations in key reward brain regions, such as medial prefrontal cortex (mPFC) and nucleus accumbens (NAc), and consequently leading to oxidative stress and neuroinflammation. Previous studies from our lab tested ß-lactam antibiotics and novel beta-lactam non-antibiotic, MC-100093, and showed these ß-lactam upregulated the major astrocytic glutamate transporter, GLT-1, and consequently reduced ethanol intake and normalized glutamate homeostasis. This present study tested the effects of novel synthetic ß-lactam non-antibiotic drug, MC-100093, in chronic ethanol intake and neuroinflammatory and trophic factors in subregions of the NAc (NAc core and shell) and mPFC (Prelimbic, PL; and Infralimbic, IL) of male P rats. MC-100093 treatment reduced ethanol intake after 5-week drinking regimen. Importantly, MC-100093 attenuated ethanol-induced downregulation of brain derived neurotrophic factor (BDNF) expression in these brain regions. In addition, MC-100093 attenuated ethanol-induced upregulation of pro-inflammatory cytokines such as TNF-a and HMGB1 in all these brain regions. Furthermore, MC-100093 treatment attenuated ethanol-induced increase in RAGE in these brain regions. MC-100093 prevented neuroinflammation caused by ethanol intake as well as increased neurotrophic factor in mesocorticolimbic brain regions. MC-100093 treatment reduced ethanol intake and this behavioral effect was associated with attenuation of reduced trophic factors and increased pro-inflammatory factors. MC-100093 is considered a small molecule that may have potential therapeutic effects for the treatment of the effects of chronic exposure to ethanol.


Asunto(s)
Etanol , Transportador 2 de Aminoácidos Excitadores , Núcleo Accumbens , Corteza Prefrontal , Animales , Masculino , Transportador 2 de Aminoácidos Excitadores/metabolismo , Etanol/farmacología , Ratas , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Biomarcadores/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Alcoholismo/tratamiento farmacológico , Alcoholismo/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos
9.
Prog Neurobiol ; 236: 102614, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38641040

RESUMEN

Complement activation and prefrontal cortical dysfunction both contribute to the pathogenesis of major depressive disorder (MDD), but their interplay in MDD is unclear. We here studied the role of complement C3a receptor (C3aR) in the medial prefrontal cortex (mPFC) and its influence on depressive-like behaviors induced by systematic lipopolysaccharides (LPS) administration. C3aR knockout (KO) or intra-mPFC C3aR antagonism confers resilience, whereas C3aR expression in mPFC neurons makes KO mice susceptible to LPS-induced depressive-like behaviors. Importantly, the excitation and inhibition of mPFC neurons have opposing effects on depressive-like behaviors, aligning with increased and decreased excitability by C3aR deletion and activation in cortical neurons. In particular, inhibiting mPFC glutamatergic (mPFCGlu) neurons, the main neuronal subpopulation expresses C3aR, induces depressive-like behaviors in saline-treated WT and KO mice, but not in LPS-treated KO mice. Compared to hypoexcitable mPFCGlu neurons in LPS-treated WT mice, C3aR-null mPFCGlu neurons display hyperexcitability upon LPS treatment, and enhanced excitation of mPFCGlu neurons is anti-depressant, suggesting a protective role of C3aR deficiency in these circumstances. In conclusion, C3aR modulates susceptibility to LPS-induced depressive-like behaviors through mPFCGlu neuronal excitability. This study identifies C3aR as a pivotal intersection of complement activation, mPFC dysfunction, and depression and a promising therapeutic target for MDD.


Asunto(s)
Depresión , Lipopolisacáridos , Ratones Noqueados , Neuronas , Corteza Prefrontal , Animales , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Lipopolisacáridos/farmacología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratones , Depresión/metabolismo , Depresión/inducido químicamente , Receptores de Complemento/metabolismo , Ratones Endogámicos C57BL , Masculino , Ácido Glutámico/metabolismo
10.
Biomed Pharmacother ; 174: 116473, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38522237

RESUMEN

BACKGROUND: The elevation of endocannabinoid levels through inhibiting their degradation afforded neuroprotection in CaMKIIα-TDP-43 mice, a conditional transgenic model of frontotemporal dementia. However, which cannabinoid receptors are mediating these benefits is still pending to be elucidated. METHODS: We have investigated the involvement of the CB1 and the CB2 receptor using chronic treatments with selective ligands in CaMKIIα-TDP-43 mice, analysis of their cognitive deterioration with the Novel Object Recognition test, and immunostaining for neuronal and glial markers in two areas of interest in frontotemporal dementia. RESULTS: Our results confirmed the therapeutic value of activating either the CB1 or the CB2 receptor, with improvements in the animal performance in the Novel Object Recognition test, preservation of pyramidal neurons, in particular in the medial prefrontal cortex, and attenuation of glial reactivity, in particular in the hippocampus. In addition, the activation of both CB1 and CB2 receptors reduced the elevated levels of TDP-43 in the medial prefrontal cortex of CaMKIIα-TDP-43 mice, an effect exerted by mechanisms that are currently under investigation. CONCLUSIONS: These data reinforce the notion that the activation of CB1 and CB2 receptors may represent a promising therapy against TDP-43-induced neuropathology in frontotemporal dementia. Future studies will have to confirm these benefits, in particular with one of the selective CB2 agonists used here, which has been thoroughly characterized for clinical development.


Asunto(s)
Cannabinoides , Modelos Animales de Enfermedad , Demencia Frontotemporal , Ratones Transgénicos , Fármacos Neuroprotectores , Receptor Cannabinoide CB1 , Receptor Cannabinoide CB2 , Animales , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/metabolismo , Masculino , Fármacos Neuroprotectores/farmacología , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB1/agonistas , Demencia Frontotemporal/tratamiento farmacológico , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Ratones , Cannabinoides/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteínas de Unión al ADN/metabolismo , Ratones Endogámicos C57BL , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología
11.
J Physiol ; 602(9): 2047-2060, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38500302

RESUMEN

Adverse experiences in early life can induce maladaptive responses to acute stress in later life. Chronic social isolation during adolescence is an early life adversity that can precipitate stress-related psychiatric disorders. We found that male mice after 8 weeks of adolescent social isolation (SI) have markedly increased aggression after being exposed to 2 h of restraint stress (RS), which was accompanied by a significant increase of AMPA receptor- and NMDA receptor-mediated synaptic transmission in prefrontal cortex (PFC) pyramidal neurons of SIRS males. Compared to group-housed counterparts, SIRS males exhibited a significantly decreased level of histone H3 acetylation in PFC. Systemic administration of class I histone deacetylase inhibitors, romidepsin or MS-275, ameliorated the aggressive behaviour, as well as general social interaction deficits, of SIRS males. Electrophysiological recordings also found normalization of PFC glutamatergic currents by romidepsin treatment of SIRS male mice. These results revealed an epigenetic mechanism and intervention avenue for aggression induced by chronic social isolation. KEY POINTS: Adolescent chronic social isolation can precipitate stress-related psychiatric disorders. A significant increase of glutamatergic transmission is found in the prefrontal cortex (PFC) of socially isolated male mice exposed to an acute stress (SIRS). Treatment with class I histone deacetylase (HDAC) inhibitors ameliorates the aggressive behaviour and social interaction deficits of SIRS males, and normalizes glutamatergic currents in PFC neurons. It provides an epigenetic mechanism and intervention avenue for aberrant stress responses induced by chronic social isolation.


Asunto(s)
Agresión , Inhibidores de Histona Desacetilasas , Ratones Endogámicos C57BL , Corteza Prefrontal , Aislamiento Social , Estrés Psicológico , Animales , Masculino , Inhibidores de Histona Desacetilasas/farmacología , Aislamiento Social/psicología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Agresión/efectos de los fármacos , Ratones , Depsipéptidos/farmacología , Piridinas/farmacología , Benzamidas/farmacología , Restricción Física , Transmisión Sináptica/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Células Piramidales/metabolismo
12.
Exp Brain Res ; 242(5): 1149-1160, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38489023

RESUMEN

Hypofunctioning of NMDA receptors, and the resulting shift in the balance between excitation and inhibition, is considered a key process in the pathophysiology of schizophrenia. One important manifestation of this phenomenon is changes in neural oscillations, those above 30 Hz (i.e., gamma-band oscillations), in particular. Although both preclinical and clinical studies observed increased gamma activity following acute administration of NMDA receptor antagonists, the relevance of this phenomenon has been recently questioned given the reduced gamma oscillations typically observed during sensory and cognitive tasks in schizophrenia. However, there is emerging, yet contradictory, evidence for increased spontaneous gamma-band activity (i.e., at rest or under baseline conditions). Here, we use the sub-chronic phencyclidine (PCP) rat model for schizophrenia, which has been argued to model the pathophysiology of schizophrenia more closely than acute NMDA antagonism, to investigate gamma oscillations (30-100 Hz) in the medial prefrontal cortex of anesthetized animals. While baseline gamma oscillations were not affected, oscillations induced by train stimulation of the posterior dorsal CA1 (pdCA1) field of the hippocampus were enhanced in PCP-treated animals (5 mg/kg, twice daily for 7 days, followed by a 7-day washout period). This effect was reversed by pharmacological enhancement of endocannabinoid levels via systemic administration of URB597 (0.3 mg/kg), an inhibitor of the catabolic enzyme of the endocannabinoid anandamide. Intriguingly, the pharmacological blockade of CB1 receptors by AM251 unmasked a reduced gamma oscillatory activity in PCP-treated animals. The findings are consistent with the observed effects of URB597 and AM251 on behavioral deficits reminiscent of the symptoms of schizophrenia and further validate the potential for cannabinoid-based drugs as a treatment for schizophrenia.


Asunto(s)
Amidohidrolasas , Benzamidas , Carbamatos , Modelos Animales de Enfermedad , Ritmo Gamma , Fenciclidina , Piperidinas , Corteza Prefrontal , Esquizofrenia , Animales , Esquizofrenia/fisiopatología , Esquizofrenia/metabolismo , Esquizofrenia/tratamiento farmacológico , Fenciclidina/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Ritmo Gamma/fisiología , Ritmo Gamma/efectos de los fármacos , Masculino , Ratas , Carbamatos/farmacología , Benzamidas/farmacología , Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/metabolismo , Piperidinas/farmacología , Pirazoles/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Endocannabinoides/metabolismo , Ácidos Araquidónicos/metabolismo , Ácidos Araquidónicos/farmacología , Ratas Sprague-Dawley , Alcamidas Poliinsaturadas/metabolismo , Alcamidas Poliinsaturadas/farmacología
13.
Neurotherapeutics ; 21(3): e00328, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38355360

RESUMEN

Methamphetamine (MA) use disorder poses significant challenges to both the affected individuals and society. Current non-drug therapies like transcranial direct-current stimulation and transcranial magnetic stimulation have limitations due to their invasive nature and limited reach to deeper brain areas. Transcranial focused ultrasound (FUS) is gaining attention as a noninvasive option with precise spatial targeting, able to affect deeper areas of the brain. This research focused on assessing the effectiveness of FUS in influencing the infralimbic cortex (IL) to prevent the recurrence of MA-seeking behavior, using the conditioned place preference (CPP) method in rats. The study involved twenty male Sprague-Dawley rats. Neuronal activation by FUS was first examined via electromyography (EMG). Rats received alternately with MA or saline, and confined to one of two distinctive compartments in a three compartment apparatus over a 4-day period. After CPP test, extinction, the first reinstatement, and extinction again, FUS was applied to IL prior to the second MA priming-induced reinstatement. Safety assessments were conducted through locomotor and histological function examinations. EMG data confirmed the effectiveness of FUS in activating neurons. Significant attenuation of reinstatement of MA CPP was found, along with successful targeting of the IL region, confirmed through acoustic field scanning, c-Fos immunohistochemistry, and Evans blue dye staining. No damage to brain tissue or impaired locomotor activity was observed. The results of the study indicate that applying FUS to the IL markedly reduced the recurrence of MA seeking behavior, without harming brain tissue or impairing motor skills. This suggests that FUS could be a promising method for treating MA use disorder, with the infralimbic cortex being an effective target for FUS in preventing MA relapse.


Asunto(s)
Extinción Psicológica , Metanfetamina , Ratas Sprague-Dawley , Animales , Masculino , Metanfetamina/farmacología , Ratas , Extinción Psicológica/efectos de los fármacos , Terapia por Ultrasonido/métodos , Estimulantes del Sistema Nervioso Central/farmacología , Corteza Prefrontal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo
14.
J Physiol ; 602(9): 2127-2139, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38285002

RESUMEN

Maternal stress and glucocorticoid exposure during pregnancy have multigenerational effects on neuroendocrine function and behaviours in offspring. Importantly, effects are transmitted through the paternal lineage. Altered phenotypes are associated with profound differences in transcription and DNA methylation in the brain. In the present study, we hypothesized that maternal prenatal synthetic glucocorticoid (sGC) exposure in the F0 pregnancy will result in differences in miRNA levels in testes germ cells and sperm across multiple generations, and that these changes will associate with modified microRNA (miRNA) profiles and gene expression in the prefrontal cortex (PFC) of subsequent generations. Pregnant guinea-pigs (F0) were treated with multiple courses of the sGC betamethasone (Beta) (1 mg kg-1; gestational days 40, 41, 50, 51, 60 and 61) in late gestation. miRNA levels were assessed in testes germ cells and in F2 PFC using the GeneChip miRNA 4.0 Array and candidate miRNA measured in epididymal sperm by quantitative real-time PCR. Maternal Beta exposure did not alter miRNA levels in germ cells derived from the testes of adult male offspring. However, there were significant differences in the levels of four candidate miRNAs in the sperm of F1 and F2 adult males. There were no changes in miRNA levels in the PFC of juvenile F2 female offspring. The present study has identified that maternal Beta exposure leads to altered miRNA levels in sperm that are apparent for at least two generations. The fact that differences were confined to epididymal sperm suggests that the intergenerational effects of Beta may target the epididymis. KEY POINTS: Paternal glucocorticoid exposure prior to conception leads to profound epigenetic changes in the brain and somatic tissues in offspring, and microRNAs (miRNAs) in sperm may mediate these changes. We show that there were significant differences in the miRNA profile of epididymal sperm in two generations following prenatal glucocorticoid exposure that were not observed in germ cells derived from the testes. The epididymis is a probable target for intergenerational programming. The effects of prenatal glucocorticoid treatment may span multiple generations.


Asunto(s)
Glucocorticoides , MicroARNs , Efectos Tardíos de la Exposición Prenatal , Espermatozoides , Animales , Femenino , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Cobayas , Glucocorticoides/farmacología , Testículo/efectos de los fármacos , Testículo/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Betametasona/farmacología , Exposición Materna/efectos adversos
15.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36835313

RESUMEN

Adolescent exposure to cannabinoids as a postnatal environmental insult may increase the risk of psychosis in subjects exposed to perinatal insult, as suggested by the two-hit hypothesis of schizophrenia. Here, we hypothesized that peripubertal Δ9-tetrahydrocannabinol (aTHC) may affect the impact of prenatal methylazoxymethanol acetate (MAM) or perinatal THC (pTHC) exposure in adult rats. We found that MAM and pTHC-exposed rats, when compared to the control group (CNT), were characterized by adult phenotype relevant to schizophrenia, including social withdrawal and cognitive impairment, as revealed by social interaction test and novel object recognition test, respectively. At the molecular level, we observed an increase in cannabinoid CB1 receptor (Cnr1) and/or dopamine D2/D3 receptor (Drd2, Drd3) gene expression in the prefrontal cortex of adult MAM or pTHC-exposed rats, which we attributed to changes in DNA methylation at key regulatory gene regions. Interestingly, aTHC treatment significantly impaired social behavior, but not cognitive performance in CNT groups. In pTHC rats, aTHC did not exacerbate the altered phenotype nor dopaminergic signaling, while it reversed cognitive deficit in MAM rats by modulating Drd2 and Drd3 gene expression. In conclusion, our results suggest that the effects of peripubertal THC exposure may depend on individual differences related to dopaminergic neurotransmission.


Asunto(s)
Dronabinol , Efectos Tardíos de la Exposición Prenatal , Esquizofrenia , Animales , Femenino , Humanos , Embarazo , Ratas , Modelos Animales de Enfermedad , Dopamina/metabolismo , Dronabinol/toxicidad , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptores de Dopamina D3/metabolismo , Esquizofrenia/inducido químicamente
16.
BMC Neurosci ; 24(1): 2, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36631757

RESUMEN

BACKGROUND: The head-twitch response (HTR) in mice is considered a behavioral model for hallucinogens and serotonin 5-HT2A receptor function, as well as Tourette syndrome in humans. It is mediated by 5-HT2A receptor agonists such as ( ±)- 2,5-dimethoxy-4-iodoamphetamine (DOI) in the prefrontal cortex (PFC). The 5-HT2A antagonist EMD 281014, can prevent both DOI-induced HTR during ageing and c-fos expression in different regions of PFC. Moreover, the nonselective monoamine releaser methamphetamine (MA) suppressed DOI-induced HTR through ageing via concomitant activation of inhibitory 5-HT1A receptors, but enhanced DOI-evoked c-fos expression. d-Fenfluramine is a selective 5-HT releaser and induces HTR in mice, whereas MA does not. Currently, we investigated whether EMD 281014 or MA would alter: (1) d-fenfluramine-induced HTR frequency in 20-, 30- and 60-day old mice, (2) d-fenfluramine-evoked c-fos expression in PFC, and (3) whether blockade of inhibitory serotonergic 5-HT1A- or adrenergic ɑ2-receptors would prevent suppressive effect of MA on d-fenfluramine-induced HTR. RESULTS: EMD 281014 (0.001-0.05 mg/kg) or MA (0.1-5 mg/kg) blocked d-fenfluramine-induced HTR dose-dependently during ageing. The 5-HT1A antagonist WAY 100635 countered the inhibitory effect of MA on d-fenfluramine-induced HTR in 30-day old mice, whereas the adrenergic ɑ2 antagonist RS 79948 reversed MA's inhibitory effect in both 20- and 30- day old mice. d-Fenfluramine significantly increased c-fos expressions in PFC regions. MA (1 mg/kg) pretreatment significantly increased d-fenfluramine-evoked c-fos expression in different regions of PFC. EMD 281014 (0.05 mg/kg) failed to prevent d-fenfluramine-induced c-fos expression, but significantly increased it in one PFC region (PrL at - 2.68 mm). CONCLUSION: EMD 281014 suppressed d-fenfluramine-induced HTR but failed to prevent d-fenfluramine-evoked c-fos expression which suggest involvement of additional serotonergic receptors in the mediation of evoked c-fos. The suppressive effect of MA on d-fenfluramine-evoked HTR is due to well-recognized functional interactions between stimulatory 5-HT2A- and the inhibitory 5-HT1A- and ɑ2-receptors. MA-evoked increases in c-fos expression in PFC regions are due to the activation of diverse monoaminergic receptors through increased synaptic concentrations of 5-HT, NE and/or DA, which may also account for the additive effect of MA on d-fenfluramine-evoked changes in c-fos expression. Our findings suggest potential drug receptor functional interaction during development when used in combination.


Asunto(s)
Fenfluramina , Metanfetamina , Corteza Prefrontal , Proteínas Proto-Oncogénicas c-fos , Animales , Humanos , Ratones , Adrenérgicos/metabolismo , Adrenérgicos/farmacología , Envejecimiento/metabolismo , Fenfluramina/metabolismo , Fenfluramina/farmacología , Metanfetamina/metabolismo , Metanfetamina/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptor de Serotonina 5-HT2A/efectos de los fármacos , Receptor de Serotonina 5-HT2A/metabolismo , Serotonina/metabolismo , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo
17.
Neurotoxicology ; 94: 126-134, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36442689

RESUMEN

Bisphenol A (BPA) is an endocrine disruptor found in polycarbonate plastics and exposure in humans is nearly ubiquitous and it has widespread effects on cognitive, emotional, and reproductive behaviors in both humans and animal models. In our laboratory we previously found that perinatal BPA exposure results in a higher number of neurons in the adult male rat prefrontal cortex (PFC) and less play in adolescents of both sexes. Here we examine changes in the rate of postnatal apoptosis in the rat prefrontal cortex and its timing with brief BPA exposure. Because an increased number of neurons in the PFC is a characteristic of a subtype of autism spectrum disorder, we tested social preference following brief BPA exposure and also expression of a small group of genes. Males and females were exposed to BPA from postnatal days (P) 6 through 8 or from P10 through 12. Both exposures significantly decreased indicators of cell death in the developing medial prefrontal cortex in male subjects only. Additionally, males exposed to BPA from P6 - 8 showed decreased social preference and decreased cortical expression of Shank3 and Homer1, two synaptic scaffolding genes that have been implicated in social deficits. There were no significant effects of BPA in the female subjects. These results draw attention to the negative consequences following brief exposure to BPA during early development.


Asunto(s)
Trastorno del Espectro Autista , Disruptores Endocrinos , Animales , Femenino , Masculino , Embarazo , Ratas , Apoptosis , Trastorno del Espectro Autista/inducido químicamente , Trastorno del Espectro Autista/metabolismo , Compuestos de Bencidrilo/toxicidad , Compuestos de Bencidrilo/metabolismo , Disruptores Endocrinos/toxicidad , Expresión Génica , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Conducta Social , Modelos Animales de Enfermedad
18.
Cereb Cortex ; 33(6): 2470-2484, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35650684

RESUMEN

The endocannabinoid (eCB) system represents a promising neurobiological target for novel anxiolytic pharmacotherapies. Previous clinical and preclinical evidence has revealed that genetic and/or pharmacological manipulations altering eCB signaling modulate fear and anxiety behaviors. Water-insoluble eCB lipid anandamide requires chaperone proteins for its intracellular transport to degradation, a process that requires fatty acid-binding proteins (FABPs). Here, we investigated the effects of a novel FABP-5 inhibitor, SBFI-103, on fear and anxiety-related behaviors using rats. Acute intra-prelimbic cortex administration of SBFI-103 induced a dose-dependent anxiolytic response and reduced contextual fear expression. Surprisingly, both effects were reversed when a cannabinoid-2 receptor (CB2R) antagonist, AM630, was co-infused with SBFI-103. Co-infusion of the cannabinoid-1 receptor antagonist Rimonabant with SBFI-103 reversed the contextual fear response yet showed no reversal effect on anxiety. Furthermore, in vivo neuronal recordings revealed that intra-prelimbic region SBFI-103 infusion altered the activity of putative pyramidal neurons in the basolateral amygdala and ventral hippocampus, as well as oscillatory patterns within these regions in a CB2R-dependent fashion. Our findings identify a promising role for FABP5 inhibition as a potential target for anxiolytic pharmacotherapy. Furthermore, we identify a novel, CB2R-dependent FABP-5 signaling pathway in the PFC capable of strongly modulating anxiety-related behaviors and anxiety-related neuronal transmission patterns.


Asunto(s)
Ansiolíticos , Ansiedad , Proteínas de Unión a Ácidos Grasos , Corteza Prefrontal , Receptor Cannabinoide CB2 , Animales , Ratas , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Ansiolíticos/metabolismo , Ansiolíticos/farmacología , Ansiolíticos/uso terapéutico , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Cannabinoides/metabolismo , Endocannabinoides/metabolismo , Proteínas de Unión a Ácidos Grasos/antagonistas & inhibidores , Proteínas de Unión a Ácidos Grasos/metabolismo , Miedo/efectos de los fármacos , Miedo/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/antagonistas & inhibidores , Receptor Cannabinoide CB2/metabolismo
19.
Cereb Cortex ; 33(8): 4806-4814, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-36156637

RESUMEN

The medial prefrontal cortex (mPFC) has been implicated in regulating resistance to the effects of acute uncontrollable stress. We previously showed that mPFC-lesioned animals exhibit impaired object recognition memory after acute exposure to a brief stress that had no effect in normal animals. Here, we used designer receptors exclusively activated by designer drugs to determine how modulating mPFC activity affects recognition-memory performance under stressful conditions. Specifically, animals with chemogenetic excitation or inhibition of the mPFC underwent either a brief ineffective stress (20-min restraint + 20 tail shocks) or a prolonged effective stress (60-min restraint + 60 tail shocks). Subsequent recognition memory tests showed that animals with chemogenetic mPFC inhibition exposed to brief stress showed impairment in an object recognition memory task, whereas those with chemogenetic mPFC excitation exposed to prolonged stress did not. Thus, the present findings the decreased mPFC activity exacerbates acute stress effects on memory function whereas increased mPFC activity counters these stress effects provide evidence that the mPFC bidirectionally modulates stress resistance.


Asunto(s)
Disfunción Cognitiva , Memoria , Corteza Prefrontal , Reconocimiento en Psicología , Estrés Fisiológico , Estrés Psicológico , Animales , Masculino , Ratas , Clozapina/análogos & derivados , Clozapina/farmacología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/prevención & control , Electrochoque/psicología , Memoria/efectos de los fármacos , Memoria/fisiología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Restricción Física/fisiología , Estrés Fisiológico/fisiología , Estrés Psicológico/complicaciones , Estrés Psicológico/fisiopatología , Factores de Tiempo
20.
Transl Psychiatry ; 12(1): 458, 2022 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-36316324

RESUMEN

Chronic morphine administration alters gene expression in different brain regions, an effect which may contribute to plastic changes associated with addictive behavior. This change in gene expression is most possibly mediated by addictive drug-induced epigenetic remodeling of gene expression programs. Our previous studies showed that chronic morphine-induced decrease of miR-105 in the medial prefrontal cortex (mPFC) contributed to context-induced retrieval of morphine withdrawal memory. However, how chronic morphine treatment decreases miR-105 in the mPFC still remains unknown. The present study shows that chronic morphine induces addiction-related change in miR-105 in the mPFC via two kinds of transcription factors: the first transcription factor is CREB activated by mu receptors-ERK-p90RSK signaling pathway and the second transcription factor is glucocorticoid receptor (GR), which as a negative transcription factor, mediates chronic morphine-induced decrease in miR-105 in the mPFC of rats.


Asunto(s)
MicroARNs , Morfina , Corteza Prefrontal , Factores de Transcripción , Animales , Ratas , Regulación de la Expresión Génica , MicroARNs/genética , Morfina/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...