Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Psychiatry ; 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879719

RESUMEN

Substance use disorders (SUD) and drug addiction are major threats to public health, impacting not only the millions of individuals struggling with SUD, but also surrounding families and communities. One of the seminal challenges in treating and studying addiction in human populations is the high prevalence of co-morbid conditions, including an increased risk of contracting a human immunodeficiency virus (HIV) infection. Of the ~15 million people who inject drugs globally, 17% are persons with HIV. Conversely, HIV is a risk factor for SUD because chronic pain syndromes, often encountered in persons with HIV, can lead to an increased use of opioid pain medications that in turn can increase the risk for opioid addiction. We hypothesize that SUD and HIV exert shared effects on brain cell types, including adaptations related to neuroplasticity, neurodegeneration, and neuroinflammation. Basic research is needed to refine our understanding of these affected cell types and adaptations. Studying the effects of SUD in the context of HIV at the single-cell level represents a compelling strategy to understand the reciprocal interactions among both conditions, made feasible by the availability of large, extensively-phenotyped human brain tissue collections that have been amassed by the Neuro-HIV research community. In addition, sophisticated animal models that have been developed for both conditions provide a means to precisely evaluate specific exposures and stages of disease. We propose that single-cell genomics is a uniquely powerful technology to characterize the effects of SUD and HIV in the brain, integrating data from human cohorts and animal models. We have formed the Single-Cell Opioid Responses in the Context of HIV (SCORCH) consortium to carry out this strategy.

2.
Nature ; 574(7778): 372-377, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31619789

RESUMEN

Diabetes is far more prevalent in smokers than non-smokers, but the underlying mechanisms of vulnerability are unknown. Here we show that the diabetes-associated gene Tcf7l2 is densely expressed in the medial habenula (mHb) region of the rodent brain, where it regulates the function of nicotinic acetylcholine receptors. Inhibition of TCF7L2 signalling in the mHb increases nicotine intake in mice and rats. Nicotine increases levels of blood glucose by TCF7L2-dependent stimulation of the mHb. Virus-tracing experiments identify a polysynaptic connection from the mHb to the pancreas, and wild-type rats with a history of nicotine consumption show increased circulating levels of glucagon and insulin, and diabetes-like dysregulation of blood glucose homeostasis. By contrast, mutant Tcf7l2 rats are resistant to these actions of nicotine. Our findings suggest that TCF7L2 regulates the stimulatory actions of nicotine on a habenula-pancreas axis that links the addictive properties of nicotine to its diabetes-promoting actions.


Asunto(s)
Trastornos del Metabolismo de la Glucosa/genética , Habénula/metabolismo , Transducción de Señal , Tabaquismo/complicaciones , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Animales , AMP Cíclico/metabolismo , Glucosa/metabolismo , Trastornos del Metabolismo de la Glucosa/metabolismo , Humanos , Ratones , Mutagénesis , Nicotina/metabolismo , Células PC12 , Páncreas/metabolismo , Ratas , Receptores Nicotínicos/metabolismo , Tabaquismo/genética , Tabaquismo/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/genética
4.
Science ; 384(6700): eadn0886, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38843332

RESUMEN

In addition to their intrinsic rewarding properties, opioids can also evoke aversive reactions that protect against misuse. Cellular mechanisms that govern the interplay between opioid reward and aversion are poorly understood. We used whole-brain activity mapping in mice to show that neurons in the dorsal peduncular nucleus (DPn) are highly responsive to the opioid oxycodone. Connectomic profiling revealed that DPn neurons innervate the parabrachial nucleus (PBn). Spatial and single-nuclei transcriptomics resolved a population of PBn-projecting pyramidal neurons in the DPn that express µ-opioid receptors (µORs). Disrupting µOR signaling in the DPn switched oxycodone from rewarding to aversive and exacerbated the severity of opioid withdrawal. These findings identify the DPn as a key substrate for the abuse liability of opioids.


Asunto(s)
Analgésicos Opioides , Reacción de Prevención , Trastornos Relacionados con Opioides , Oxicodona , Núcleos Parabraquiales , Corteza Prefrontal , Receptores Opioides mu , Recompensa , Animales , Masculino , Ratones , Analgésicos Opioides/farmacología , Conectoma , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Trastornos Relacionados con Opioides/metabolismo , Oxicodona/farmacología , Núcleos Parabraquiales/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Células Piramidales/metabolismo , Receptores Opioides mu/metabolismo , Receptores Opioides mu/genética , Síndrome de Abstinencia a Sustancias/metabolismo , Transcriptoma
5.
Int J Neuropsychopharmacol ; 16(8): 1885-92, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23684180

RESUMEN

Early-life stress is a main contributory factor to the onset of depression. Treatments remain inadequate and as such, a large unmet medical need for novel therapeutics remains. Impeding advancement is the poor understanding of the molecular pathology. microRNAs (miRNAs) are novel regulators of gene expression. A paucity of information regarding their role in depressive pathology and antidepressant action remains. This study investigated changes to hippocampal miRNA levels induced via early-life stress in Sprague-Dawley rats and whether antidepressant treatments could reverse these changes. Investigated were the selective serotonin reuptake inhibitor fluoxetine, the rapid acting N-methyl-d-aspartate receptor antagonist ketamine and electroconvulsive shock therapy (ECT). Microarray analysis revealed early-life stress affected the expression of multiple hippocampal miRNAs. Antidepressant treatments reversed some of these effects including a stress-induced change to miR-451. Ketamine and ECT possessed the highest number of common targets suggesting convergence on common pathways. Interestingly all three treatments possessed miR-598-5p as a common target. This demonstrates that changes to hippocampal miRNA expression may represent an important component of stress-induced pathology and antidepressant action may reverse these.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/terapia , Terapia Electroconvulsiva/métodos , Ketamina/uso terapéutico , MicroARNs/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Biología Computacional , Depresión/etiología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Masculino , Privación Materna , MicroARNs/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas
6.
Behav Pharmacol ; 24(2): 105-13, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23455446

RESUMEN

There is increasing evidence suggesting a role of the neurotransmitter glutamate in depression. The metabotropic glutamate (mGlu) receptors are G-protein coupled receptors, which mediate a slow modulatory response to glutamate signalling. mGlu7 receptor is a presynaptic inhibitory autoreceptor showing great promise as a potential therapeutic target for the treatment of depression. Selective pharmacological modulators of mGlu7 receptor have been developed; the positive allosteric modulator AMN082 and the negative modulator 6-(4-methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one hydrochloride (MMPIP). They remain to be extensively characterized in behavioural models sensitive to antidepressant action. Therefore, we assessed the effects of these compounds on behaviour in two different mouse strains using several preclinical tests sensitive to antidepressant pharmacological action. AMN082 (6 mg/kg) reduced immobility in the forced swim test and tail suspension test (TST) in both C57BL/6j and CD1 mice. In CD1 mice, MMPIP (10 and 30 mg/kg) significantly increased the time spent immobile in the TST, whereas this effect was restricted to a dose of 30 mg/kg in C57BL/6j mice. Administration of MMPIP with AMN082 partially attenuated the antidepressant-like effect of AMN082 in C57BL/6j mice in the forced swim test and the TST. However, this effect was absent from the CD1 strain. This further adds to the growing corpus of data promoting the targeting of mGlu7 receptor with the aim of achieving an antidepressant effect.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Terapia Molecular Dirigida , Receptores de Glutamato Metabotrópico/agonistas , Animales , Animales no Consanguíneos , Antidepresivos/administración & dosificación , Antidepresivos/antagonistas & inhibidores , Conducta Animal/efectos de los fármacos , Compuestos de Bencidrilo/administración & dosificación , Compuestos de Bencidrilo/antagonistas & inhibidores , Compuestos de Bencidrilo/uso terapéutico , Depresión/inducido químicamente , Depresión/metabolismo , Relación Dosis-Respuesta a Droga , Antagonismo de Drogas , Evaluación Preclínica de Medicamentos , Suspensión Trasera , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Piridonas/administración & dosificación , Piridonas/efectos adversos , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/metabolismo , Especificidad de la Especie , Natación
7.
Artículo en Inglés | MEDLINE | ID: mdl-35636576

RESUMEN

Rates of obesity and obesity-associated diseases have increased dramatically in countries with developed economies. Substance use disorders (SUDs) are characterized by the persistent use of the substance despite negative consequences. It has been hypothesized that overconsumption of palatable energy dense food can elicit SUD-like maladaptive behaviors that contribute to persistent caloric intake beyond homeostatic need even in the face of negative consequences. Palatable food and drugs of abuse act on many of the same motivation-related circuits in the brain, and can induce, at least superficially, similar molecular, cellular, and physiological adaptations on these circuits. As such, applying knowledge about the neurobiological mechanisms of SUDs may serve as useful heuristic to better understand the persistent overconsumption of palatable food that contributes to obesity. However, many important differences exist between the actions of drugs of abuse and palatable food in the brain. This warrants caution when attributing weight gain and obesity to the manifestation of a putative SUD-related behavioral disorder. Here, we describe similarities and differences between compulsive drug use in SUDs and overconsumption in obesity and consider the merit of the concept of "food addiction".


Asunto(s)
Heurística , Trastornos Relacionados con Sustancias , Conducta Alimentaria/fisiología , Humanos , Hiperfagia , Obesidad
8.
Gastroenterology ; 138(7): 2418-25, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20226190

RESUMEN

BACKGROUND & AIMS: The molecular basis underlying visceral hypersensitivity in functional irritable bowel syndrome remains elusive, resulting in poor treatment effectiveness. Because alterations in spinal non-neuronal (astrocytic) glutamate reuptake are suspected to participate in chronic pain, we asked whether such processes occur in visceral hypersensitivity. METHODS: Visceral hypersensitivity was induced in Sprague-Dawley rats by maternal separation. Separated adults were given a systemic administration of riluzole (5 mg/kg), an approved neuroprotective agent activating glutamate reuptake. Visceral hypersensitivity was assessed using colorectal distension (40 mm Hg). Somatic nociception was quantified using Hot Plate, Randall-Sellito, and Hargreaves tests. Spinal proteins were quantified using immunofluorescence and Western blot. The dependence of visceral sensory function upon spinal glutamate transport was evaluated by intrathecal injection of glutamate transport antagonist DL-threo-beta-benzyloxyaspartate (TBOA). For in vitro testing of riluzole and TBOA, primary cultures of astrocytes were used. RESULTS: We show that riluzole counteracts stress-induced visceral hypersensitivity without affecting visceral response in nonseparated rats or altering nociceptive responses to somatic pain stimulation. In addition, maternal separation produces a reduction in glial excitatory amino acid transporter (EAAT)-1 with no change in EAAT-2 or gamma-amino butyric acid transporters. Stress was not associated with changes in glial fibrillary acidic protein or astrocytic morphology per se. Furthermore, visceral normosensitivity relies on spinal EAAT, as intrathecal TBOA is sufficient to induce hypersensitivity in normal rats. CONCLUSIONS: We identify spinal EAAT as a therapeutic target, and establish riluzole as a candidate to counteract gastrointestinal hypersensitivity in disorders such as irritable bowel syndrome.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/uso terapéutico , Ácido Glutámico/metabolismo , Síndrome del Colon Irritable/tratamiento farmacológico , Riluzol/uso terapéutico , Médula Espinal/metabolismo , Estrés Psicológico/complicaciones , Animales , Ácido Aspártico/farmacología , Transportador 1 de Aminoácidos Excitadores/análisis , Transportador 1 de Aminoácidos Excitadores/fisiología , Transportador 2 de Aminoácidos Excitadores/análisis , Transportador 2 de Aminoácidos Excitadores/fisiología , Proteína Ácida Fibrilar de la Glía/análisis , Masculino , Privación Materna , Actividad Motora , Ratas , Ratas Sprague-Dawley
9.
Nat Commun ; 12(1): 5121, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433818

RESUMEN

Comparatively little is known about how new instrumental actions are encoded in the brain. Using whole-brain c-Fos mapping, we show that neural activity is increased in the anterior dorsolateral striatum (aDLS) of mice that successfully learn a new lever-press response to earn food rewards. Post-learning chemogenetic inhibition of aDLS disrupts consolidation of the new instrumental response. Similarly, post-learning infusion of the protein synthesis inhibitor anisomycin into the aDLS disrupts consolidation of the new response. Activity of D1 receptor-expressing medium spiny neurons (D1-MSNs) increases and D2-MSNs activity decreases in the aDLS during consolidation. Chemogenetic inhibition of D1-MSNs in aDLS disrupts the consolidation process whereas D2-MSN inhibition strengthens consolidation but blocks the expression of previously learned habit-like responses. These findings suggest that D1-MSNs in the aDLS encode new instrumental actions whereas D2-MSNs oppose this new learning and instead promote expression of habitual actions.


Asunto(s)
Condicionamiento Operante , Cuerpo Estriado/fisiología , Neuronas/fisiología , Animales , Conducta Animal , Cuerpo Estriado/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Long-Evans , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo
10.
Science ; 368(6487): 197-201, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32273471

RESUMEN

Vulnerability to relapse during periods of attempted abstinence from cocaine use is hypothesized to result from the rewiring of brain reward circuitries, particularly ventral tegmental area (VTA) dopamine neurons. How cocaine exposures act on midbrain dopamine neurons to precipitate addiction-relevant changes in gene expression is unclear. We found that histone H3 glutamine 5 dopaminylation (H3Q5dop) plays a critical role in cocaine-induced transcriptional plasticity in the midbrain. Rats undergoing withdrawal from cocaine showed an accumulation of H3Q5dop in the VTA. By reducing H3Q5dop in the VTA during withdrawal, we reversed cocaine-mediated gene expression changes, attenuated dopamine release in the nucleus accumbens, and reduced cocaine-seeking behavior. These findings establish a neurotransmission-independent role for nuclear dopamine in relapse-related transcriptional plasticity in the VTA.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Trastornos Relacionados con Cocaína/psicología , Cocaína/efectos adversos , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Comportamiento de Búsqueda de Drogas , Histonas/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Trastornos Relacionados con Cocaína/genética , Regulación de la Expresión Génica , Glutamina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica
11.
Psychopharmacology (Berl) ; 236(1): 265-272, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30215216

RESUMEN

RATIONALE: Post-traumatic stress disorder (PTSD) is a devastating anxiety-related disorder which develops subsequent to a severe psychologically traumatic event. Only ~ 9% of people who experience such a trauma develop PTSD. It is clear that a number of factors, including genetics, influence whether an individual will develop PTSD subsequent to a trauma. The 129S1/SvImJ (S1) inbred mouse strain displays poor fear extinction and may be useful to model this specific aspect of PTSD. The metabotropic glutamate receptor 7 (mGlu7 receptor) has previously been shown to be involved in cognitive processes and anxiety-like behaviour placing it in a key position to regulate fear extinction processes. We sought to compare mGlu7 receptor mRNA levels in the S1 strain with those in the robustly extinguishing C57BL/6J (B6) inbred strain using in situ hybridisation (ISH) in three brain regions associated with fear extinction: the amygdala, hippocampus and prefrontal cortex (PFC). RESULTS: Compared to the B6 strain, S1 mice had increased mGlu7 receptor mRNA levels in the lateral amygdala (LA) and basolateral amygdala (BLA) subdivisions. An increase was also seen in the hippocampal CA1 and CA3 subregions of S1 mice. No difference in mGlu7 receptor levels were seen in the central nucleus (CeA) of the amygdala, dentate gyrus (DG) of the hippocampus or prefrontal cortex. CONCLUSIONS: These data show altered mGlu7 receptor expression in key brain regions associated with fear extinction in two different inbred mouse strains which differ markedly in their fear extinction behaviour. Altered mGlu7 receptor levels may contribute to the deficit fear extinction processes seen in fear extinction in the S1 strain.


Asunto(s)
Modelos Animales de Enfermedad , Extinción Psicológica/fisiología , Miedo/fisiología , ARN Mensajero/biosíntesis , Receptores de Glutamato Metabotrópico/biosíntesis , Amígdala del Cerebelo/metabolismo , Animales , Trastornos de Ansiedad/genética , Trastornos de Ansiedad/metabolismo , Miedo/psicología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Corteza Prefrontal/metabolismo , ARN Mensajero/genética , Receptores de Glutamato Metabotrópico/genética
12.
Curr Drug Targets ; 17(5): 538-616, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25777273

RESUMEN

Extensive research over the past thirty years has demonstrated a vital role for metabotropic glutamate (mGlu) receptors in the major functions of the central nervous system (CNS). A wealth of preclinical studies provide evidence that pharmacological targeting of mGlu receptors can effectively attenuate the development of symptoms and progression of many CNS disorders in animal models. In this review we summarize the current knowledge on the involvement of mGlu receptors in the pathophysiology of neuropsychiatric disorders (schizophrenia, depression, anxiety and cognitive disorders, pain perception and addiction), as well as neurodegenerative (Alzheimer's, Huntington's and Parkinson's diseases) and neurodevelopmental (fragile X syndrome and autism spectrum disorders) diseases. We further emphasize the therapeutic potential of mGlu receptors' pharmacological modulators in these diseases, describe the results of clinical trials with these compounds and discuss the potential sources of translational difficulties.


Asunto(s)
Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Enfermedades del Sistema Nervioso Central/metabolismo , Ensayos Clínicos como Asunto , Humanos , Receptores de Glutamato Metabotrópico/agonistas , Transducción de Señal/efectos de los fármacos
13.
Trends Pharmacol Sci ; 37(12): 1029-1044, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27832923

RESUMEN

Current treatment strategies for psychiatric disorders remain inadequate. Impeding development of novel therapeutics is our incomplete knowledge of the molecular pathophysiology underlying these disorders. Changes to miRNA function and expression are increasingly being associated with pathological behavioral states. Furthermore, the prospect of using of miRNA expression profiles (the miRNome) as objective psychiatric diagnosis tools is gaining traction. In this review, we focus on recent findings surrounding the link between miRNA function and psychiatric disorders, and outline some of the key challenges that will need to be overcome if the therapeutic potential of these molecular effectors is to be fully realized.


Asunto(s)
Perfilación de la Expresión Génica , Trastornos Mentales/fisiopatología , MicroARNs/genética , Animales , Regulación de la Expresión Génica , Humanos , Trastornos Mentales/genética , Trastornos Mentales/terapia
15.
Neuropharmacology ; 99: 168-76, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26187394

RESUMEN

Adolescence marks a critical time when the brain is highly susceptible to pathological insult yet also uniquely amenable to therapeutic intervention. It is during adolescence that the onset of the majority of psychiatric disorders, including substance use disorder (SUDs), occurs. It has been well established that stress, particularly during early development, can contribute to the pathological changes which contribute to the development of SUDs. Glutamate as the main excitatory neurotransmitter in the mammalian CNS plays a key role in various physiological processes, including reward function, and in mediating the effects of psychological stress. We hypothesised impairing glutamatergic signalling during the key adolescent period would attenuate early-life stress induced impaired reward function. To test this, we induced early-life stress in male rats using the maternal-separation procedure. During the critical adolescent period (PND25-46) animals were treated with the glutamate transporter activator, riluzole, or the NMDA receptor antagonist, memantine. Adult reward function was assessed using voluntary cocaine intake measured via intravenous self-administration. We found that early-life stress in the form of maternal-separation impaired reward function, reducing the number of successful cocaine-infusions achieved during the intravenous self-administration procedure as well impairing drug-induced reinstatement of cocaine-taking behaviour. Interestingly, riluzole and memantine treatment reversed this stress-induced impairment. These data suggest that reducing glutamatergic signalling may be a viable therapeutic strategy for treating vulnerable individuals at risk of developing SUDs including certain adolescent populations, particularly those which may have experienced trauma during early-life.


Asunto(s)
Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Recompensa , Estrés Psicológico/metabolismo , Administración Intravenosa , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Trastornos Relacionados con Cocaína/metabolismo , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico , Masculino , Privación Materna , Memantina/farmacología , Ratas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Riluzol/farmacología , Autoadministración , Estrés Psicológico/tratamiento farmacológico
16.
Pharmacol Res Perspect ; 3(6): e00190, 2015 12.
Artículo en Inglés | MEDLINE | ID: mdl-27022464

RESUMEN

Recent preclinical studies have revealed a functionally important role for the drug efflux pump P-glycoprotein (P-gp) at the blood-brain barrier in limiting brain levels and thus antidepressant-like activity of certain antidepressant drugs. Specifically, acute administration of P-gp inhibitors, such as verapamil and cyclosporin A (CsA), has been shown to augment brain concentrations and functional activity of the antidepressant escitalopram in rodents. However, depression is a chronic disorder and current treatments require prolonged administration to elicit their full therapeutic effect. Thus, it is important to investigate whether acute findings in relation to P-gp inhibition translate to chronic paradigms. To this end, the present study investigates whether chronic treatment with the P-gp inhibitor verapamil and the antidepressant escitalopram results in enhanced brain distribution and antidepressant-like effects of escitalopram. Verapamil (10 mg·kg(-1) i.p.) and escitalopram (0.1 mg·kg(-1) i.p.) were administered once daily for 22 days. On the final day of treatment, brain regions and plasma were collected for analysis of cortical and plasma escitalopram concentrations, and to determine the hippocampal expression of genes previously reported to be altered by chronic antidepressant treatment. Verapamil treatment resulted in a greater than twofold increase in brain levels of escitalopram, without altering plasma levels. Neither gene expression analysis nor behavioral testing revealed an augmentation of responses to escitalopram treatment due to verapamil administration. Taken together, these data demonstrate for the first time that P-gp inhibition can yield elevated brain concentrations of an antidepressant after chronic treatment. The functional relevance of these increased brain levels requires further elaboration.

17.
Neurobiol Stress ; 2: 28-33, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26844237

RESUMEN

Glutamate, the main excitatory neurotransmitter in the central nervous system, exerts its effect through ionotropic and metabotropic receptors. Of these, group III mGlu receptors (mGlu 4, 6, 7, 8) are among the least studied due to a lack of pharmacological tools. mGlu7 receptors, the most highly conserved isoform, are abundantly distributed in the brain, especially in regions, such as the amygdala, known to be crucial for the emotional processing of painful stimuli. Visceral hypersensitivity is a poorly understood phenomenon manifesting as an increased sensitivity to visceral stimuli. Glutamate has long been associated with somatic pain processing leading us to postulate that crossover may exist between these two modalities. Moreover, stress has been shown to exacerbate visceral pain. ADX71743 is a novel, centrally penetrant, negative allosteric modulator of mGlu7 receptors. Thus, we used this tool to explore the possible involvement of this receptor in the mediation of visceral pain in a stress-sensitive model of visceral hypersensitivity, namely the Wistar Kyoto (WKY) rat. ADX71743 reduced visceral hypersensitivity in the WKY rat as exhibited by increased visceral sensitivity threshold with concomitant reductions in total number of pain behaviours. Moreover, AD71743 increased total distance and distance travelled in the inner zone of the open field. These findings show, for what is to our knowledge, the first time, that mGlu7 receptor signalling plays a role in visceral pain processing. Thus, negative modulation of the mGlu7 receptor may be a plausible target for the amelioration of stress-induced visceral pain where there is a large unmet medical need.

18.
Pharmacol Biochem Behav ; 123: 55-76, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24161683

RESUMEN

While antidepressant drugs are beneficial to many patients, current treatments for depression remain sub-optimal. Up to half of patients with a major depressive episode fail to achieve remission with a first line antidepressant treatment. Identification of the molecular mechanisms that dictate whether a patient will successfully respond to a particular antidepressant treatment while tolerating its side-effects is not only a major challenge in biological psychiatry research but is also one that shows great promise. This review summarises data from both clinical and preclinical studies that point to a role of specific genes in the response and resistance to antidepressant therapeutics. Moreover, we discuss how such findings have increased our understanding of the mechanism of action of antidepressant drugs. Finally, we comment on how this information may potentially influence the future development of personalised medicine approaches for the treatment of depression.


Asunto(s)
Antidepresivos/farmacología , Farmacogenética , Animales , Antidepresivos/farmacocinética , Humanos , Ratones
19.
Behav Brain Res ; 261: 17-25, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24280122

RESUMEN

Recent studies have raised the possibility that P-glycoprotein (P-gp) inhibition may represent a putative augmentation strategy for treatment with certain antidepressants. Indeed, we have previously shown that administration of the P-gp inhibitor verapamil increased the brain distribution and behavioural effects of the antidepressant escitalopram. The aim of the current study was to investigate if similar effects occur with another P-gp inhibitor, cyclosporin A (CsA). CsA pre-treatment exacerbated the severity of behaviours in an escitalopram-induced mouse model of serotonin syndrome, a potentially life-threatening adverse drug reaction associated with serotonergic drugs. P-gp inhibition by CsA enhanced the brain distribution of escitalopram by 70-80%. Serotonin (5-HT) turnover in the prefrontal cortex was reduced by escitalopram, and this effect was augmented by CsA. However, CsA pre-treatment did not augment the effect of escitalopram in the tail suspension test (TST) of antidepressant-like activity. Microdialysis experiments revealed that pre-treatment with CsA failed to augment, but blunted, the increase in extracellular 5-HT in response to escitalopram administration. This blunting effect may contribute to the lack of augmentation in the TST. Taken together, the present studies demonstrate that co-administration of CsA and escitalopram produces differential effects depending on the behavioural and neurochemical assays employed. Thus, the results highlight the need for further studies involving more selective pharmacological tools to specifically evaluate the impact of P-gp inhibition on behavioural responses to antidepressants which are subject to efflux by P-gp.


Asunto(s)
Antidepresivos/toxicidad , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Citalopram/toxicidad , Ciclosporina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Síndrome de la Serotonina/inducido químicamente , Síndrome de la Serotonina/tratamiento farmacológico , 5-Hidroxitriptófano/toxicidad , Animales , Encéfalo/metabolismo , Ciclosporina/farmacología , Modelos Animales de Enfermedad , Combinación de Medicamentos , Electroquímica , Inhibidores Enzimáticos/farmacología , Suspensión Trasera/métodos , Ácido Hidroxiindolacético/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
20.
Eur J Pharmacol ; 698(1-3): 19-30, 2013 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-23123053

RESUMEN

L-glutamate is produced by a great variety of peripheral tissues in both health and disease. Like other components of the glutamatergic system, metabotropic glutamate (mGlu) receptors also have a widespread distribution outside the central nervous system (CNS). In particular, group III mGlu receptors have been recently found in human stomach and colon revealing an extraordinary potential for these receptors in the treatment of peripheral disorders, including gastrointestinal dysfunction. The significance of these findings is that pharmacological tools originally designed for mGlu receptors in the CNS may also be directed towards new disease targets in the periphery. Targeting mGlu receptors can also be beneficial in the treatment of disorders involving central components together with gastrointestinal dysfunction, such as irritable bowel syndrome, which can be co-morbid with anxiety and depression. Conversely, the development of more specific therapeutic approaches for mGlu ligands both centrally as in the gut will depend on the elucidation of tissue-specific elements in mGlu receptor signalling.


Asunto(s)
Encéfalo/metabolismo , Tracto Gastrointestinal/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Enfermedad , Humanos , Sistema Nervioso Periférico/metabolismo , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA