Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.201
Filtrar
1.
Theranostics ; 14(7): 2881-2896, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38773977

RESUMEN

Methamphetamine (METH) withdrawal anxiety symptom and relapse have been significant challenges for clinical practice, however, the underlying neuronal basis remains unclear. Our recent research has identified a specific subpopulation of choline acetyltransferase (ChAT+) neurons localized in the external lateral portion of parabrachial nucleus (eLPBChAT), which modulates METH primed-reinstatement of conditioned place preference (CPP). Here, the anatomical structures and functional roles of eLPBChAT projections in METH withdrawal anxiety and primed reinstatement were further explored. Methods: In the present study, a multifaceted approach was employed to dissect the LPBChAT+ projections in male mice, including anterograde and retrograde tracing, acetylcholine (Ach) indicator combined with fiber photometry recording, photogenetic and chemogenetic regulation, as well as electrophysiological recording. METH withdrawal anxiety-like behaviors and METH-primed reinstatement of conditioned place preference (CPP) were assessed in male mice. Results: We identified that eLPBChAT send projections to PKCδ-positive (PKCδ+) neurons in lateral portion of central nucleus of amygdala (lCeAPKCδ) and oval portion of bed nucleus of the stria terminalis (ovBNSTPKCδ), forming eLPBChAT-lCeAPKCδ and eLPBChAT-ovBNSTPKCδ pathways. At least in part, the eLPBChAT neurons positively innervate lCeAPKCδ neurons and ovBNSTPKCδ neurons through regulating synaptic elements of presynaptic Ach release and postsynaptic nicotinic acetylcholine receptors (nAChRs). METH withdrawal anxiety and METH-primed reinstatement of CPP respectively recruit eLPBChAT-lCeAPKCδ pathway and eLPBChAT-ovBNSTPKCδ pathway in male mice. Conclusion: Our findings put new insights into the complex neural networks, especially focusing on the eLPBChAT projections. The eLPBChAT is a critical node in the neural networks governing METH withdrawal anxiety and primed-reinstatement of CPP through its projections to the lCeAPKCδ and ovBNSTPKCδ, respectively.


Asunto(s)
Ansiedad , Metanfetamina , Ratones Endogámicos C57BL , Síndrome de Abstinencia a Sustancias , Animales , Metanfetamina/efectos adversos , Masculino , Ratones , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología , Ansiedad/metabolismo , Neuronas/metabolismo , Colina O-Acetiltransferasa/metabolismo , Núcleos Septales/metabolismo , Conducta Animal/efectos de los fármacos
2.
Neurosci Lett ; 832: 137804, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38692559

RESUMEN

The present study aimed to investigate the role of agmatine in the neurobiology underlying memory impairment during ethanol withdrawal in rats. Sprague-Dawley rats were subjected to a 21-day chronic ethanol exposure regimen (2.4 % w/v ethanol for 3 days, 4.8 % w/v for the next 4 days, and 7.2 % w/v for the following 14 days), followed by a withdrawal period. Memory impairment was assessed using the passive avoidance test (PAT) at 24, 48, and 72 h post-withdrawal. The ethanol-withdrawn rats displayed a significant decrease in step-through latency in the PAT, indicative of memory impairment at 72 h post-withdrawal. However, administration of agmatine (40 µg/rat) and its modulators (L-arginine, arcaine, and amino-guanidine) significantly increases the latency time in the ethanol-withdrawn rats, demonstrating the attenuation of memory impairment. Further, pretreatment with imidazoline receptor agonists enhances agmatine's effects, while antagonists block them, implicating imidazoline receptors in agmatine's actions. Neurochemical analysis in ethanol-withdrawn rats reveals dysregulated glutamate and GABA levels, which was attenuated by agmatine and its modulators. By examining the effects of agmatine administration and modulators of endogenous agmatine, the study aimed to shed light on the potential therapeutic implications of agmatinergic signaling in alcohol addiction and related cognitive deficits. Thus, the present findings suggest that agmatine administration and modulation of endogenous agmatine levels hold potential as therapeutic strategies for managing alcohol addiction and associated cognitive deficits. Understanding the neurobiology underlying these effects paves the way for the development of novel interventions targeting agmatinergic signaling in addiction treatment.


Asunto(s)
Agmatina , Disfunción Cognitiva , Etanol , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias , Animales , Agmatina/farmacología , Agmatina/uso terapéutico , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/psicología , Masculino , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Ratas , Biguanidas/farmacología , Ácido Glutámico/metabolismo , Arginina/farmacología , Ácido gamma-Aminobutírico/metabolismo , Receptores de Imidazolina/metabolismo , Receptores de Imidazolina/agonistas , Reacción de Prevención/efectos de los fármacos
3.
Brain Res ; 1835: 148930, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38604556

RESUMEN

The anxiety caused by morphine protracted abstinence is considered to be an important factor contributes to drug-seeking and relapse. Endoplasmic reticulum (ER) stress plays important roles in many kinds of mental disorders including drug addiction and anxiety, but it is unclear whether ER stress is involved in anxiety-like behaviors induced by morphine withdrawal. In this study, by using behavioral test, western blot, immunofluorescence, electron transmission microscope, we found that: (1) Inhibition of endoplasmic reticulum stress by 4-Phenylbutyric acid (4-PBA) could attenuate anxiety-like behaviors induced by morphine withdrawal. (2) The endoplasmic reticulum stress-related proteins in the lateral habenula (LHb) but not in the nucleus accumbens (NAc), ventral pallidum (VP), basolateral amygdala (BLA) and CA1 of hippocampus was upregulated by morphine withdrawal, upregulation of endoplasmic reticulum stress-related proteins in the lateral habenula induced by morphine withdrawal was inhibited by 4-PBA. (3) Endoplasmic reticulum stress-related protein CHOP and eIF2α were expressed in neurons but not in microglia in the LHb. (4) Morphine withdrawal induced neuronal morphological change in the LHb, which was attenuated by 4-PBA.


Asunto(s)
Ansiedad , Estrés del Retículo Endoplásmico , Morfina , Síndrome de Abstinencia a Sustancias , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Masculino , Morfina/farmacología , Ansiedad/metabolismo , Ansiedad/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo , Ratones , Fenilbutiratos/farmacología , Dependencia de Morfina/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratones Endogámicos C57BL
4.
Drug Alcohol Depend ; 259: 111276, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38676968

RESUMEN

BACKGROUND: As nicotine dependence represents a longstanding major public health issue, new nicotine cessation pharmacotherapies are needed. Administration of N-oleoyl glycine (OlGly), an endogenous lipid signaling molecule, prevents nicotine-induced conditioned place preference (CPP) through a peroxisome proliferator-activated receptor-alpha (PPARα) dependent mechanism, and also ameliorated withdrawal signs in nicotine-dependent mice. Pharmacological evidence suggests that the methylated analog of OlGly, N-oleoyl alanine (OlAla), has an increased duration of action and may offer translational benefit. Accordingly, OlAla was assessed in nicotine CPP and dependence assays as well as its pharmacokinetics compared to OlGly. METHODS: ICR female and male mice were tested in nicotine-induced CPP with and without the PPARα antagonist GW6471. OlAla was also assessed in nicotine-dependent mice following removal of nicotine minipumps: somatic withdrawal signs, thermal hyper-nociception and altered affective behavior (i.e., light/dark box). Finally, plasma and brain were collected after administration of OlGly or OlAla and analyzed by high-performance liquid chromatography tandem mass spectrometry. RESULTS: OlAla prevented nicotine-induced CPP, but this effect was not blocked by GW6471. OlAla attenuated somatic and affective nicotine withdrawal signs, but not thermal hyper-nociception in nicotine-dependent mice undergoing withdrawal. OlAla and OlGly showed similar time-courses in plasma and brain. CONCLUSIONS: The observation that both molecules showed similar pharmacokinetics argues against the notion that OlAla offers increased metabolic stability. Moreover, while these structurally similar lipids show efficacy in mouse models of reward and dependence, they reduce nicotine reward through distinct mechanisms.


Asunto(s)
Ratones Endogámicos ICR , Nicotina , Recompensa , Síndrome de Abstinencia a Sustancias , Tabaquismo , Animales , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Ratones , Masculino , Nicotina/farmacología , Femenino , Tabaquismo/metabolismo , PPAR alfa/metabolismo , Alanina/farmacología , Alanina/análogos & derivados , Ácidos Oléicos/farmacología , Glicina/farmacología , Glicina/análogos & derivados , Aminopiridinas/farmacología , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Oxazoles , Tirosina/análogos & derivados
5.
Pharmacol Res ; 202: 107143, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38499081

RESUMEN

Substance use disorders (SUD) are chronic relapsing disorders governed by continually shifting cycles of positive drug reward experiences and drug withdrawal-induced negative experiences. A large body of research points to plasticity within systems regulating emotional, motivational, and cognitive processes as drivers of continued compulsive pursuit and consumption of substances despite negative consequences. This plasticity is observed at all levels of analysis from molecules to networks, providing multiple avenues for intervention in SUD. The cytoskeleton and its regulatory proteins within neurons and glia are fundamental to the structural and functional integrity of brain processes and are potentially the major drivers of the morphological and behavioral plasticity associated with substance use. In this review, we discuss preclinical studies that provide support for targeting the brain cytoskeleton as a therapeutic approach to SUD. We focus on the interplay between actin cytoskeleton dynamics and exposure to cocaine, methamphetamine, alcohol, opioids, and nicotine and highlight preclinical studies pointing to a wide range of potential therapeutic targets, such as nonmuscle myosin II, Rac1, cofilin, prosapip 1, and drebrin. These studies broaden our understanding of substance-induced plasticity driving behaviors associated with SUD and provide new research directions for the development of SUD therapeutics.


Asunto(s)
Síndrome de Abstinencia a Sustancias , Trastornos Relacionados con Sustancias , Humanos , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Trastornos Relacionados con Sustancias/metabolismo , Citoesqueleto , Citoesqueleto de Actina/metabolismo , Encéfalo , Síndrome de Abstinencia a Sustancias/metabolismo
6.
Psychopharmacology (Berl) ; 241(6): 1265-1275, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38396195

RESUMEN

There is a significant co-occurrence of opioid use disorder (OUD) and post-traumatic stress disorder (PTSD) in clinical populations. However, the neurobiological mechanisms linking chronic opioid use, withdrawal, and the development of PTSD are poorly understood. Our previous research has shown that proinflammatory cytokines, expressed primarily by astrocytes in the dorsal hippocampus (DH), play a role in the development of heroin withdrawal-enhanced fear learning (HW-EFL), an animal model of PTSD-OUD comorbidity. Given the role of astrocytes in memory, fear learning, and opioid use, our experiments aimed to investigate their involvement in HW-EFL. Experiment 1 examined the effect of withdrawal from chronic heroin administration on GFAP surface area and volume, and identified increased surface area and volume of GFAP immunoreactivity in the dentate gyrus (DG) following 24-hour heroin withdrawal. Experiment 2 examined astrocyte morphology and synaptic interactions at the 24-hour withdrawal timepoint using an astroglial membrane-bound GFP (AAV5-GfaABC1D-lck-GFP). Although we did not detect significant changes in surface area and volume of GfaABC1D-Lck-GFP labelled astrocytes, we did observe a significant increase in the colocalization of astrocyte membranes with PSD-95 (postsynaptic density protein 95) in the DG. Experiment 3 tested if stimulating astroglial Gi signaling in the DH alters HW-EFL, and our results demonstrate this manipulation attenuates HW-EFL. Collectively, these findings contribute to our current understanding of the effects of heroin withdrawal on astrocytes and support the involvement of astrocytes in the comorbid relationship between opioid use and anxiety disorders.


Asunto(s)
Astrocitos , Miedo , Heroína , Hipocampo , Síndrome de Abstinencia a Sustancias , Astrocitos/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Heroína/administración & dosificación , Masculino , Hipocampo/metabolismo , Miedo/fisiología , Trastornos por Estrés Postraumático/metabolismo , Aprendizaje/fisiología , Modelos Animales de Enfermedad , Dependencia de Heroína/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Ratones
7.
Neuropharmacology ; 247: 109846, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38211698

RESUMEN

Tobacco smoking remains a leading cause of preventable death in the United States, with approximately a 5% success rate for smokers attempting to quit. High relapse rates have been linked to several genetic factors, indicating that the mechanistic relationship between genes and drugs of abuse is a valuable avenue for the development of novel smoking cessation therapies. For example, various single nucleotide polymorphisms (SNPs) in the gene for neuregulin 3 (NRG3) and its cognate receptor, the receptor tyrosine-protein kinase erbB-4 (ERBB4), have been linked to nicotine addiction. Our lab has previously shown that ERBB4 plays a role in anxiety-like behavior during nicotine withdrawal (WD); however, the neuronal mechanisms and circuit-specific effects of NRG3-ERBB4 signaling during nicotine and WD are unknown. The present study utilizes genetic, biochemical, and functional approaches to examine the anxiety-related behavioral and functional role of NRG3-ERBB4 signaling, specifically in the ventral hippocampus (VH) of male and female mice. We report that 24hWD from nicotine is associated with altered synaptic expression of VH NRG3 and ERBB4, and genetic disruption of VH ErbB4 leads to an elimination of anxiety-like behaviors induced during 24hWD. Moreover, we observed attenuation of GABAergic transmission as well as alterations in Ca2+-dependent network activity in the ventral CA1 area of VH ErbB4 knock-down mice during 24hWD. Our findings further highlight contributions of the NRG3-ERBB4 signaling pathway to anxiety-related behaviors seen during nicotine WD.


Asunto(s)
Nicotina , Síndrome de Abstinencia a Sustancias , Masculino , Femenino , Ratones , Animales , Nicotina/farmacología , Nicotina/metabolismo , Neurregulinas/genética , Neurregulinas/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Hipocampo/metabolismo , Transducción de Señal , Receptor ErbB-4/genética , Receptor ErbB-4/metabolismo
8.
Biol Psychiatry ; 95(3): 275-285, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37562519

RESUMEN

BACKGROUND: The ventral tegmental area (VTA) is a dopaminergic brain area that is critical in the development and maintenance of addiction. During withdrawal from chronic ethanol exposure, the response of VTA neurons to GABA (gamma-aminobutyric acid) is reduced through an epigenetically regulated mechanism. In the current study, a whole-genome transcriptomic approach was used to investigate the underlying molecular mechanism of GABA hyposensitivity in the VTA during withdrawal after chronic ethanol exposure. METHODS: We performed RNA sequencing of the VTA of Sprague Dawley male rats withdrawn for 24 hours from a chronic ethanol diet as well as sequencing of the VTA of control rats fed the Lieber-DeCarli diet. RNA sequencing data were analyzed using weighted gene coexpression network analysis to identify modules that contained coexpressed genes. Validation was performed with quantitative polymerase chain reaction, gas chromatography-mass spectrometry, and electrophysiological assays. RESULTS: Pathway and network analysis of weighted gene coexpression network analysis module 1 revealed a significant downregulation of genes associated with the cholesterol synthesis pathway. Consistent with this association, VTA cholesterol levels were significantly decreased during withdrawal. Chromatin immunoprecipitation indicated a decrease in levels of acetylated H3K27 at the transcriptional control regions of these genes. Electrophysiological studies in VTA slices demonstrated that GABA hyposensitivity during withdrawal was normalized by addition of exogenous cholesterol. In addition, inhibition of cholesterol synthesis produced GABA hyposensitivity, which was reversed by adding exogenous cholesterol to VTA slices. CONCLUSIONS: These results suggest that decreased expression of cholesterol synthesis genes may regulate GABA hyposensitivity of VTA neurons during alcohol withdrawal. Increasing cholesterol levels in the brain may be a novel avenue for therapeutic intervention to reverse detrimental effects of chronic alcohol exposure.


Asunto(s)
Alcoholismo , Síndrome de Abstinencia a Sustancias , Ratas , Masculino , Animales , Ácido gamma-Aminobutírico/metabolismo , Síndrome de Abstinencia a Sustancias/genética , Síndrome de Abstinencia a Sustancias/metabolismo , Área Tegmental Ventral , Alcoholismo/metabolismo , Ratas Sprague-Dawley , Etanol/farmacología
9.
Neuron ; 112(1): 113-123.e4, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37909038

RESUMEN

Rehabilitation from alcohol addiction or abuse is hampered by withdrawal symptoms including severe headaches, which often lead to rehabilitation failure. There is no appropriate therapeutic option available for alcohol-withdrawal-induced headaches. Here, we show the role of the mast-cell-specific receptor MrgprB2 in the development of alcohol-withdrawal-induced headache. Withdrawing alcohol from alcohol-acclimated mice induces headache behaviors, including facial allodynia, facial pain expressions, and reduced movement, which are symptoms often observed in humans. Those behaviors were absent in MrgprB2-deficient mice during alcohol withdrawal. We observed in vivo spontaneous activation and hypersensitization of trigeminal ganglia (TG) neurons in alcohol-withdrawal WT mice, but not in alcohol-withdrawal MrgprB2-deficient mice. Increased mast cell degranulation by alcohol withdrawal in dura mater was dependent on the presence of MrgprB2. The results indicate that alcohol withdrawal causes headache via MrgprB2 of mast cells in dura mater, suggesting that MrgprB2 is a potential target for treating alcohol-withdrawal-related headaches.


Asunto(s)
Alcoholismo , Síndrome de Abstinencia a Sustancias , Humanos , Ratones , Masculino , Animales , Mastocitos/metabolismo , Síndrome de Abstinencia a Sustancias/complicaciones , Síndrome de Abstinencia a Sustancias/metabolismo , Ganglio del Trigémino/fisiología , Cefalea/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
10.
J Agric Food Chem ; 71(49): 19531-19550, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38038704

RESUMEN

Increasing evidence points to the critical role of calcium overload triggered by mitochondrial dysfunction in the development of alcoholic liver disease (ALD). As an important organelle for aerobic respiration with a double-layered membrane, mitochondria are pivotal targets of alcohol metabolism-mediated lipid peroxidation, wherein mitochondria-specific phospholipid cardiolipin oxidation to 4-hydroxynonenal (4-HNE) ultimately leads to mitochondrial integrity and function impairment. Therefore, it is absolutely essential to identify effective nutritional intervention targeting mitochondrial redox function for an alternative therapy of ALD, in order to compensate for the difficulty in achieving alcohol withdrawal due to addiction. In this study, we confirmed the significant advantages of astaxanthin (AX) against alcohol toxicity among various carotenoids via cell experiments and identified the potential in mitochondrion morphogenesis and calcium signaling pathway by bioinformatics analysis. The ALD model of Sprague-Dawley (SD) rats was also generated to investigate the effectiveness of AX on alcohol-induced liver injury, and the underlying mechanisms were further explored. AX intervention attenuated alcohol-induced oxidative stress and lipid peroxidation as well as mitochondrial dysfunction characterized by degenerative morphology changes and collapsed membrane potential. Also, AX reduced the production of 4-HNE by activating the Nrf2-ARE signaling pathway, which is closely associated with the redox balance of mitochondria. In addition, relieved mitochondrial Ca2+ accumulation caused by AX was observed both in vivo and in vitro. Furthermore, we revealed the structure-activity relationship of AX and mitochondrial membrane channel proteins MCU and VDAC1, implying potential acting targets. Altogether, our data indicated a new mechanism of AX intervention which protects against alcohol-induced liver injury through restoring redox balance and Ca2+ homeostasis in mitochondria, as well as provided novel insights into the development of AX as a therapeutic option for the management of ALD.


Asunto(s)
Alcoholismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Hepatopatías Alcohólicas , Enfermedades Mitocondriales , Síndrome de Abstinencia a Sustancias , Ratas , Animales , Calcio/metabolismo , Alcoholismo/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Hígado/metabolismo , Estrés Oxidativo , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/prevención & control , Hepatopatías Alcohólicas/metabolismo , Etanol/metabolismo , Proteínas de la Membrana/metabolismo , Enfermedades Mitocondriales/metabolismo , Homeostasis
11.
Neuropharmacology ; 240: 109732, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37774943

RESUMEN

Prenatal opioid exposure is a major health concern in the United States, with the incidence of neonatal opioid withdrawal syndrome (NOWS) escalating in recent years. NOWS occurs upon cessation of in utero opioid exposure and is characterized by increased irritability, disrupted sleep patterns, high-pitched crying, and dysregulated feeding. The main pharmacological strategy for alleviating symptoms is treatment with replacement opioids. The neural mechanisms mediating NOWS and the long-term neurobehavioral effects are poorly understood. We used a third trimester-approximate model in which neonatal outbred pups (Carworth Farms White; CFW) were administered once-daily morphine (15 mg/kg, s.c.) from postnatal day (P) day 1 through P14 and were then assessed for behavioral and transcriptomic adaptations within the nucleus accumbens (NAc) on P15. We also investigated the long-term effects of perinatal morphine exposure on adult learning and reward sensitivity. We observed significant weight deficits, spontaneous thermal hyperalgesia, and altered ultrasonic vocalization (USV) profiles following repeated morphine and during spontaneous withdrawal. Transcriptome analysis of NAc from opioid-withdrawn P15 neonates via bulk mRNA sequencing identified an enrichment profile consistent with downregulation of myelin-associated transcripts. Despite the neonatal behavioral and molecular effects, there were no significant long-term effects of perinatal morphine exposure on adult spatial memory function in the Barnes Maze, emotional learning in fear conditioning, or in baseline or methamphetamine-potentiated reward sensitivity as measured via intracranial self-stimulation. Thus, the once daily third trimester-approximate exposure regimen, while inducing NOWS model traits and significant transcriptomic effects in neonates, had no significant long-term effects on adult behaviors.


Asunto(s)
Síndrome de Abstinencia Neonatal , Trastornos Relacionados con Opioides , Síndrome de Abstinencia a Sustancias , Embarazo , Femenino , Animales , Ratones , Analgésicos Opioides/farmacología , Núcleo Accumbens , Vaina de Mielina , Síndrome de Abstinencia a Sustancias/metabolismo , Narcóticos/farmacología , Morfina/farmacología , Síndrome de Abstinencia Neonatal/tratamiento farmacológico , Síndrome de Abstinencia Neonatal/epidemiología , Síndrome de Abstinencia Neonatal/etiología , Expresión Génica , Trastornos Relacionados con Opioides/metabolismo
12.
Neurobiol Learn Mem ; 205: 107831, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37730099

RESUMEN

The cognitive effects of nicotine are linked to persistent modifications in extended neural systems that regulate cognitive and emotional processes, and these changes occur during development. Additionally, acute stress has modulatory effects on cognition that involve broad neural systems and can be influenced by prior environmental challenges. The effects of nicotine and stress may be interconnected, leading to modifications in a network of shared brain substrates. Here, we explored the interaction between nicotine and stress by evaluating the effects of acute stress exposure in spatial memory retrieval for animals pretreated with nicotine during adolescence or adulthood. Adolescent (35 days old) and adult (70 days old) male Wistar rats were treated for 21 days with one daily subcutaneous injection of nicotine 0.14 mg/ml (free base). 30 days after the last injection, rats were trained in the Barnes maze and tested 24 h later, half the rats were tested under regular conditions, and half of them were exposed to 1 h of restraining stress before the retrieval test, and brain samples were collected and c-Fos immunopositive cells were stained. Prolonged nicotine withdrawal or acute stress improved spatial memory retrieval. Acute stress in nicotine pretreated adults impaired spatial memory retrieval. Nicotine exposure during early adulthood resulted in long-lasting brain adaptations that amplified emotional responses to acute stress after prolonged drug withdrawal.


Asunto(s)
Nicotina , Síndrome de Abstinencia a Sustancias , Ratas , Masculino , Animales , Nicotina/farmacología , Memoria Espacial , Ratas Wistar , Encéfalo/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo
13.
Nutrients ; 15(13)2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37447362

RESUMEN

Multiple alcohol use disorder (AUD)-related behavioral alterations are governed by protein kinase C epsilon (PKCε), particularly in the amygdala. Protein kinase C (PKC) is readily phosphorylated at Ser729 before activation by the mTORC2 protein complex. In keeping with this, the current study was conducted to assess the variations in mTORC2 and PKCε during different ethanol exposure stages. The following groups of rats were employed: control, acute, chronic, ethanol withdrawal (EW), and EW + ethanol (EtOH). Ethanol-containing and non-ethanol-containing modified liquid diets (MLDs) were administered for 27 days. On day 28, either saline or ethanol (2.5 g/kg, 20% v/v) was intraperitoneally administered, followed by bilateral amygdala extraction. PKCε mRNA levels were noticeably increased in the amygdala of the EW + EtOH and EW groups. Following chronic ethanol consumption, the stress-activated map kinase-interacting protein 1 (Sin1) gene expression was markedly decreased. In the EW, EW + EtOH, and chronic ethanol groups, there was a profound increase in the protein expression of mTOR, Sin1, PKCε, and phosphorylated PKCε (Ser729). The PKCε gene and protein expressions showed a statistically significant moderate association, according to a correlation analysis. Our results suggest that an elevated PKCε protein expression in the amygdala during EW and EW + EtOH occurred at the transcriptional level. However, an elevation in the PKCε protein expression, but not its mRNA, after chronic ethanol intake warrants further investigation to fully understand the signaling pathways during different episodes of AUD.


Asunto(s)
Alcoholismo , Síndrome de Abstinencia a Sustancias , Ratas , Animales , Alcoholismo/metabolismo , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo , Roedores , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Etanol , Amígdala del Cerebelo , Síndrome de Abstinencia a Sustancias/metabolismo , ARN Mensajero/metabolismo
14.
Drug Alcohol Depend ; 248: 109938, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37267743

RESUMEN

The rising prevalence of early-life opioid exposure has become a pressing public health issue in the U.S. Neonates exposed to opioids in utero are at risk of experiencing a constellation of postpartum withdrawal symptoms commonly referred to as neonatal opioid withdrawal syndrome (NOWS). Buprenorphine (BPN), a partial agonist at the mu-opioid receptor (MOR) and antagonist at the kappa-opioid receptor (KOR), is currently approved to treat opioid use disorder in adult populations. Recent research suggests that BPN may also be effective in reducing withdrawal symptoms in neonates who were exposed to opioids in utero. We sought to determine whether BPN attenuates somatic withdrawal in a mouse model of NOWS. Our findings indicate that the administration of morphine (10mg/kg, s.c.) from postnatal day (PND) 1-14 results in increased somatic symptoms upon naloxone-precipitated (1mg/kg, s.c.) withdrawal. Co-administration of BPN (0.3mg/kg, s.c.) from PND 12-14 attenuated symptoms in morphine-treated mice. On PND 15, 24h following naloxone-precipitated withdrawal, a subset of mice was examined for thermal sensitivity in the hot plate test. BPN treatment significantly increased response latency in morphine-exposed mice. Lastly, neonatal morphine exposure elevated mRNA expression of KOR, and reduced mRNA expression of corticotropin-releasing hormone (CRH) in the periaqueductal gray when measured on PND 14. Altogether, this data provides support for the therapeutic effects of acute low-dose buprenorphine treatment in a mouse model of neonatal opioid exposure and withdrawal.


Asunto(s)
Buprenorfina , Síndrome de Abstinencia a Sustancias , Femenino , Ratones , Animales , Buprenorfina/farmacología , Buprenorfina/uso terapéutico , Morfina/farmacología , Analgésicos Opioides/farmacología , Naloxona/farmacología , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo , Receptores Opioides , ARN Mensajero , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/uso terapéutico
15.
Brain Res ; 1813: 148428, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37263551

RESUMEN

Protracted opioid withdrawal is considered to be a traumatic event with many adverse effects. However, little attention is paid to its consequences on the protein expression in the rat brain. A better understanding of the changes at the molecular level is essential for designing future innovative drug therapies. Our previous proteomic data indicated that long-term morphine withdrawal is associated with altered proteins functionally involved in energy metabolism, cytoskeletal changes, oxidative stress, apoptosis, or signal transduction. In this study, we selected peroxiredoxin II (PRX II) as a marker of oxidative stress, 14-3-3 proteins as adaptors, and creatine kinase-B (CK-B) as a marker of energy metabolism to detect their amounts in the brain cortex and hippocampus isolated from rats after 3-month (3 MW) and 6-month morphine withdrawal (6 MW). Methodically, our work was based on immunoblotting accompanied by 2D resolution of PRX II and 14-3-3 proteins. Our results demonstrate significant upregulation of PRX II in the rat brain cortex (3-fold) and hippocampus (1.3-fold) after 3-month morphine abstinence, which returned to the baseline six months since the drug was withdrawn. Interestingly, the level of 14-3-3 proteins was downregulated in both brain areas in 3 MW samples and remained decreased only in the brain cortex of 6 MW. Our findings suggest that the rat brain cortex and hippocampus exhibit the oxidative stress-induced vulnerability represented by compensatory upregulation of PRX II after three months of morphine withdrawal.


Asunto(s)
Dependencia de Morfina , Síndrome de Abstinencia a Sustancias , Ratas , Animales , Morfina/metabolismo , Proteínas 14-3-3/metabolismo , Regulación hacia Arriba , Proteómica , Peroxirredoxinas/metabolismo , Peroxirredoxinas/farmacología , Hipocampo/metabolismo , Encéfalo/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo
16.
Artículo en Inglés | MEDLINE | ID: mdl-37348641

RESUMEN

Although the negative effects coming along with opiate withdrawal are in part modulated by L-type calcium channels (LTCCs), the distinctive physiological properties and functions of LTCCs subtypes suggest differential roles of subtypes during withdrawal. The present study aimed to examine the contributions of LTCC subtypes, Cav1.2 and Cav1.3, within the dorsal hippocampus (DH) in naloxone-precipitated morphine withdrawal using the conditioned place aversion (CPA) paradigm. Firstly, we injected the non-specific LTCCs antagonist verapamil into the DH of morphine-dependent rats before conditioning an environment with naloxone-precipitated withdrawal. Our results showed that verapamil blocked the acquisition of CPA. Then, to explore the molecular mechanisms of LTCCs subtypes during withdrawal, we measured the protein expression of Cav1.2 and Cav1.3 in morphine-dependent rats under different conditions. In morphine-dependent rats, conditioning with withdrawal increased Cav1.2 expression in the membrane, while only acute naloxone injection increased the membrane expression of Cav1.3. To further determine the causal roles of LTCCs subtypes in the withdrawal process, we used Cav1.2 siRNA or Cav1.3 shRNA to knock down the expression of subtypes and detected the effects on CPA and somatic withdrawal signs in morphine-dependent rats. Cav1.2 siRNA, but not Cav1.3 shRNA, inhibited the acquirement of CPA and relieved somatic withdrawal symptoms. Together, our findings reveal that Cav1.2, but not Cav1.3 plays an important role in mediating morphine withdrawal, suggesting this subtype may serve as a potential therapeutic target for the treatment of negative effects in opiate dependence.


Asunto(s)
Dependencia de Morfina , Síndrome de Abstinencia a Sustancias , Ratas , Animales , Morfina/farmacología , Canales de Calcio Tipo L , Naloxona/farmacología , Dependencia de Morfina/metabolismo , Hipocampo/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Verapamilo/farmacología , Antagonistas de Narcóticos/farmacología , Reacción de Prevención
17.
Addict Biol ; 28(5): e13276, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37186439

RESUMEN

Calcium/calmodulin-dependent kinase II (CaMKII) is a key enzyme at the glutamatergic synapses. CAMK2A gene variants have been linked with alcohol use disorder (AUD) by an unknown mechanism. Here, we looked for the link between αCaMKII autophosphorylation and the AUD aetiology. Autophosphorylation-deficient heterozygous αCaMKII mutant mice (T286A+/- ) were trained in the IntelliCages to test the role of αCaMKII activity in AUD-related behaviours. The glutamatergic synapses morphology in CeA was studied in the animals drinking alcohol using 3D electron microscopy. We found that T286A+/- mutants consumed less alcohol and were more sensitive to sedating effects of alcohol, as compared to wild-type littermates (WT). After voluntary alcohol drinking, T286A+/- mice had less excitatory synapses in the CeA, as compared to alcohol-naive animals. This change correlated with alcohol consumption was not reversed after alcohol withdrawal and not observed in WT mice. Our study suggests that αCaMKII autophosphorylation affects alcohol consumption by controlling sedative effects of alcohol and preventing synaptic loss in the individuals drinking alcohol. This finding advances our understanding of the molecular processes that regulate alcohol dependence.


Asunto(s)
Alcoholismo , Síndrome de Abstinencia a Sustancias , Animales , Ratones , Alcoholismo/genética , Alcoholismo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Etanol/farmacología , Etanol/metabolismo , Fosforilación/genética , Síndrome de Abstinencia a Sustancias/metabolismo , Sinapsis/metabolismo
18.
Behav Brain Res ; 450: 114504, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37209879

RESUMEN

The misuse of and addiction to opioids are serious public health problems in some countries, such as the USA. Drug addiction is a chronic and relapsing medical condition that involves motivational and memory-related processes due to the strong associations between drugs and consuming-related stimuli. These stimuli usually trigger continuous and compulsive use and are associated with relapses after periods of withdrawal. Several factors contribute to relapse, including withdrawal-induced mood changes. Therefore, drugs attenuating withdrawal-induced affective alterations could be useful alternative treatments for relapse prevention. Cannabidiol (CBD), a non-psychotomimetic component from the Cannabis sativa plant, has anti-anxiety and anti-stress properties and has been investigated as an alternative for the treatment of several mental disorders, including drug addiction. Here, we evaluated if CBD administered 30 min prior to test for a conditioned place aversion (CPA) would attenuate the aversion induced by morphine withdrawal precipitated by the opioid receptor antagonist naloxone in male C57BL/6 mice. We also investigated if this effect involves the activation of 5-HT1A receptors, a mechanism previously associated with CBD anti-aversive effects. As expected, morphine-treated mice spent less time exploring the compartment paired with the naloxone-induced withdrawal, indicating a CPA induced by naloxone-precipitated morphine withdrawal. This effect was not observed in animals treated with CBD, at 30 and 60 mg/kg, prior to the CPA test, indicating that CBD attenuated the expression of CPA induced by naloxone-precipitated morphine withdrawal. Pretreatment with the 5-HT1A receptor antagonist WAY100635 (0.3 mg/kg) blocked CBD effects. Our findings suggest that CBD may reduce the expression of a previously established conditioned aversion induced by morphine withdrawal by a mechanism involving the activation of 5-HT1A receptors. Thus, CBD may be a therapeutic alternative for preventing relapse to opioid addiction by decreasing withdrawal-induced negative affective changes.


Asunto(s)
Cannabidiol , Dependencia de Morfina , Síndrome de Abstinencia a Sustancias , Ratones , Animales , Naloxona/farmacología , Morfina/efectos adversos , Cannabidiol/farmacología , Receptor de Serotonina 5-HT1A , Reacción de Prevención , Síndrome de Abstinencia a Sustancias/metabolismo , Ratones Endogámicos C57BL , Antagonistas de Narcóticos/farmacología , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/metabolismo
19.
Neuropsychopharmacology ; 48(11): 1567-1578, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37059867

RESUMEN

The role of lysophosphatidic acid (LPA) signaling in psychiatric disorders and drug abuse is significant. LPA receptors are widely expressed in the central nervous system, including the lateral habenula (LHb). Recent studies suggest that LHb is involved in a negative emotional state during alcohol withdrawal, which can lead to relapse. The current study examines the role of LHb LPA signaling in the negative affective state associated with alcohol withdrawal. Adult male Long-Evans rats were trained to consume either alcohol or water for eight weeks. At 48 h of withdrawal, alcohol-drinking rats showed anxiety- and depression-like symptoms, along with a significant increase in LPA signaling and related neuronal activation molecules, including autotaxin (ATX, Enpp2), LPA receptor 1/3 (LPA1/3), ßCaMKII, and c-Fos. However, there was a decrease in lipid phosphate phosphatase-related protein type 4 (LPPR4) in the LHb. Intra-LHb infusion of the LPA1/3 receptor antagonist ki-16425 or PKC-γ inhibitor Go-6983 reduced the abnormal behaviors and elevated relapse-like ethanol drinking. It also normalized high LPA1/3 receptors and enhanced AMPA GluA1 phosphorylation in Ser831 and GluA1/GluA2 ratio. Conversely, selective activation of LPA1/3 receptors by intra-LHb infusion of 18:1 LPA induced negative affective states and upregulated ßCaMKII-AMPA receptor phosphorylation in Naive rats, which were reversed by pretreatment with intra-LHb Go-6983. Our findings suggest that disturbances in LPA signaling contribute to adverse affective disorders during alcohol withdrawal, likely through PKC-γ/ßCaMKII-linked glutamate signaling. Targeting LPA may therefore be beneficial for individuals suffering from alcohol use disorders.


Asunto(s)
Alcoholismo , Habénula , Síndrome de Abstinencia a Sustancias , Humanos , Ratas , Masculino , Animales , Alcoholismo/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Habénula/metabolismo , Ratas Long-Evans
20.
Neuropsychopharmacology ; 48(9): 1318-1327, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37041207

RESUMEN

After drug withdrawal, a key factor triggering relapse is progressively intensified cue-associated drug craving, termed incubation of drug craving. After withdrawal from cocaine self-administration, incubation of cocaine craving develops more reliably in rats compared to mice. This species difference provides an opportunity to determine rat-specific cellular adaptations, which may constitute the critical mechanisms that contribute to incubated cocaine craving in humans. Expression of incubated cocaine seeking is mediated, in part, by cocaine-induced cellular adaptations in medium spiny neurons (MSNs) within the nucleus accumbens (NAc). In rats, decreased membrane excitability in NAc MSNs is a prominent cellular adaptation, which is induced after cocaine self-administration and lasts throughout prolonged drug withdrawal. Here, we show that, similar to rats, mice exhibit decreased membrane excitability of dopamine D1 receptor (D1)-, but not D2 (D2)-, expressing MSNs within the NAc shell (NAcSh) after 1 d withdrawal from cocaine self-administration. However, in contrast to rats, this membrane adaptation does not persist in mice, diminishing after 45-d withdrawal. We also find that restoring the membrane excitability of NAcSh MSNs after cocaine withdrawal decreases cocaine seeking in rats. This suggests that drug-induced membrane adaptations are essential for behavioral expression of incubated cocaine craving. In mice, however, experimentally inducing hypoactivity of D1 NAcSh MSNs after cocaine withdrawal does not alter cocaine seeking, suggesting that MSN hypo-excitability alone is insufficient to increase cocaine seeking. Together, our results demonstrate an overall permissive role of cocaine-induced hypoactivity of NAcSh MSNs in gating increased cocaine seeking after prolonged cocaine withdrawal.


Asunto(s)
Trastornos Relacionados con Cocaína , Cocaína , Síndrome de Abstinencia a Sustancias , Humanos , Ratas , Ratones , Animales , Ansia , Núcleo Accumbens/metabolismo , Neuronas/metabolismo , Trastornos Relacionados con Cocaína/metabolismo , Receptores Dopaminérgicos/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA