Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 151(1): 11-27, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31361031

RESUMEN

Adenosine receptor subtypes, first described 40 years ago, are known to regulate diverse biological functions and have a role in various conditions, such as cerebral and cardiac ischemia, immune and inflammatory disorders and cancer. In the brain, they limit potentially dangerous over excitation, but also regulate mechanisms essential in sleep and psychiatric disorders. In this review, we discuss the role of adenosine receptors in mood and anxiety disorders. Activation of A2A receptors is associated with increased depression-like symptoms, while increased A1 receptors signaling elicits rapid antidepressant effects. Indeed, several lines of evidence demonstrate that the therapeutic effects of different non-pharmacological treatments of depression, like sleep deprivation and electroconvulsive therapy are mediated by A1 receptor up-regulation or activation. In addition, A1 receptors may also play a role in the antidepressant effects of transcranial direct current stimulation and deep brain stimulation. As a potential downstream mechanism, which facilitates the antidepressant effects of A1 receptors, we propose a crosstalk between adenosinergic and glutamatergic systems mediated via synaptic plasticity protein Homer1a and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. Moreover, adenosine receptors are also involved in the control of circadian rhythms, sleep homeostasis and some neuro-immunological mechanisms, all of them implicated in mood regulation. Antagonists of adenosine receptors such as caffeine have general anxiogenic effects. In particular, A2A receptors appear to have an important role in the pathophysiology of anxiety disorders. Taken together, the results discussed here indicate that the adenosinergic system is involved in both the etiology and the treatment of mood and anxiety disorders.


Asunto(s)
Trastornos de Ansiedad/metabolismo , Trastornos del Humor/metabolismo , Receptores Purinérgicos P1/metabolismo , Animales , Humanos
2.
Glia ; 64(1): 76-89, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26295445

RESUMEN

Recently, we have demonstrated that ramified microglia are neuroprotective in N-methyl-D-aspartate (NMDA)-induced excitotoxicity in organotypic hippocampal slice cultures (OHSCs). The present study aimed to elucidate the underlying neuron-glia communication mechanism. It is shown here that pretreatment of OHSC with high concentrations of adenosine 5'-triphosphate (ATP) reduced NMDA-induced neuronal death only in presence of microglia. Specific agonists and antagonists identified the P2X7 receptor as neuroprotective receptor which was confirmed by absence of ATP-dependent neuroprotection in P2X7-deficient OHSC. Microglia replenished chimeric OHSC consisting of wild-type tissue replenished with P2X7-deficient microglia confirmed the involvement of microglial P2X7 receptor in neuroprotection. Stimulation of P2X7 in primary microglia induced tumor necrosis factor α (TNFα) release and blocking TNFα by a neutralizing antibody in OHSC abolished neuroprotection by ATP. OHSC from TNFα-deficient mice show increased exicitoxicity and activation of P2X7 did not rescue neuronal survival in the absence of TNFα. The neuroprotective effect of valproic acid (VPA) was strictly dependent on the presence of microglia and was mediated by upregulation of P2X7 in the cells. The present study demonstrates that microglia-mediated neuroprotection depends on ATP-activated purine receptor P2X7 and induction of TNFα release. This neuroprotective pathway was strengthened by VPA elucidating a novel mechanism for the neuroprotective function of VPA.


Asunto(s)
Microglía/fisiología , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Receptores Purinérgicos P2X7/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ácido Valproico/farmacología , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/patología , N-Metilaspartato/metabolismo , N-Metilaspartato/toxicidad , Neuronas/efectos de los fármacos , Neuronas/patología , ARN Mensajero/metabolismo , Receptores Purinérgicos P2X7/genética , Técnicas de Cultivo de Tejidos , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba/efectos de los fármacos
3.
Biol Chem ; 397(3): 207-14, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26641965

RESUMEN

Homer1a is upregulated by several different antidepressant measures, including non-pharmacological treatments, like sleep deprivation (SD) and electroconvulsive therapy (ECT) and antidepressant drugs, such as imipramine, fluoxetine and ketamine. Homer1a induction might thus be a crucial joint mechanism for antidepressant therapy in general. However, the upstream signaling pathways that regulate or induce Homer1a expression are still not well understood. The main focus of the present review is to offer an overview of the current knowledge about the potential role of Homer1a in depression and the signaling pathways responsible for Homer1a regulation. It is suggested here that a detailed characterization of the signaling mechanisms leading to Homer1a expression might provide novel therapeutic targets for antidepressant drug development.


Asunto(s)
Proteínas Portadoras/genética , Depresión/metabolismo , Regulación de la Expresión Génica , Transducción de Señal , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Depresión/tratamiento farmacológico , Depresión/genética , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/genética , Trastorno Depresivo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Andamiaje Homer , Humanos , Receptor de Adenosina A1/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas ras/metabolismo
4.
Glia ; 62(4): 592-607, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24470356

RESUMEN

ATP is an important regulator of microglia and its effects on microglial cytokine release are currently discussed as important contributors in a variety of brain diseases. We here analyzed the effects of ATP on the production of six inflammatory mediators (IL-6, IL-10, CCL2, IFN-γ, TNF-α, and IL-12p70) in cultured mouse primary microglia. Stimulation of P2X7 receptor by ATP (1 mM) or BzATP (500 µM) evoked the mRNA expression and release of proinflammatory cytokines IL-6, TNF-α, and the chemokine CCL2 in WT cells but not in P2X7(-/-) cells. The effects of ATP and BzATP were inhibited by the nonselective P2 receptor antagonists PPADs and suramin. Various selective P2X7 receptor antagonists blocked the P2X7-dependent release of IL-6 and CCL2, but, surprisingly, had no effect on BzATP-induced release of TNF-α in microglia. Calcium measurements confirmed that P2X7 is the main purine receptor activated by BzATP in microglia and showed that all P2X7 antagonists were functional. It is also presented that pannexin-1 hemichannel function and potential P2X4/P2X7 heterodimers are not involved in P2X7-dependent release of IL-6, CCL2, and TNF-α in microglia. How P2X7-specific antagonists only affect P2X7-dependent IL-6 and CCL2 release, but not TNF-α release is at the moment unclear, but indicates that the P2X7-dependent release of cytokines in microglia is differentially regulated.


Asunto(s)
Diferenciación Celular/fisiología , Quimiocina CCL2/metabolismo , Interleucina-6/metabolismo , Microglía/fisiología , Receptores Purinérgicos P2X7/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Animales Recién Nacidos , Encéfalo/citología , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/genética , Conexinas/genética , Conexinas/metabolismo , Relación Dosis-Respuesta a Droga , Interleucina-6/genética , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Agonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X4/deficiencia , Receptores Purinérgicos P2X7/deficiencia , Receptores Purinérgicos P2X7/genética , Factor de Necrosis Tumoral alfa/genética
5.
Neurosci Biobehav Rev ; 126: 23-42, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33757815

RESUMEN

Psychiatric disorders exhibit an enormous burden on the health care systems worldwide accounting for around one-third of years lost due to disability among adults. Their etiology is largely unknown and diagnostic classification is based on symptomatology and course of illness and not on objective biomarkers. Most psychiatric disorders are moderately to highly heritable. However, it is still unknown what mechanisms may explain the discrepancy between heritability estimates and the present data from genetic analysis. In addition to genetic differences also epigenetic modifications are considered as potentially relevant in the transfer of susceptibility to psychiatric diseases. Though, whether or not epigenetic alterations can be inherited for many generations is highly controversial. In the present article, we will critically summarize both the genetic findings and the results from epigenetic analyses, including also those of noncoding RNAs. We will argue that one possible solution to the "missing heritability" problem in psychiatry is a potential role of retrotransposons, the exploration of which is presently only in its beginnings.


Asunto(s)
Trastornos Mentales , Psiquiatría , Elementos Transponibles de ADN/genética , Epigénesis Genética , Epigenómica , Humanos , Trastornos Mentales/genética
6.
Neuropharmacology ; 162: 107834, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682853

RESUMEN

Resilience to stress is critical for the development of depression. Enhanced adenosine A1 receptor (A1R) signaling mediates the antidepressant effects of acute sleep deprivation (SD). However, chronic SD causes long-lasting upregulation of brain A1R and increases the risk of depression. To investigate the effects of A1R on mood, we utilized two transgenic mouse lines with inducible A1R overexpression in forebrain neurons. These two lines have identical levels of A1R increase in the cortex, but differ in the transgenic A1R expression in the hippocampus. Switching on the transgene promotes robust antidepressant and anxiolytic effects in both lines. The mice of the line without transgenic A1R overexpression in the hippocampus (A1Hipp-) show very strong resistance towards development of stress-induced chronic depression-like behavior. In contrast, the mice of the line in which A1R upregulation extends to the hippocampus (A1Hipp+), exhibit decreased resilience to depression as compared to A1Hipp-. Similarly, automatic analysis of reward behavior of the two lines reveals that depression resistant A1Hipp-transgenic mice exhibit high sucrose preference, while mice of the vulnerable A1Hipp + line developed stress-induced anhedonic phenotype. The A1Hipp + mice have increased Homer1a expression in hippocampus, correlating with impaired long-term potentiation in the CA1 region, mimicking the stressed mice. Furthermore, virus-mediated overexpression of Homer1a in the hippocampus decreases stress resilience. Taken together our data indicate for first time that increased expression of A1R and Homer1a in the hippocampus modulates the resilience to stress-induced depression and thus might potentially mediate the detrimental effects of chronic sleep restriction on mood.


Asunto(s)
Corteza Cerebral/metabolismo , Depresión/genética , Hipocampo/metabolismo , Proteínas de Andamiaje Homer/genética , Receptor de Adenosina A1/genética , Resiliencia Psicológica , Privación de Sueño/metabolismo , Estrés Psicológico/genética , Animales , Conducta Animal , Región CA1 Hipocampal/metabolismo , Depresión/metabolismo , Depresión/psicología , Prueba de Laberinto Elevado , Potenciales Postsinápticos Excitadores , Suspensión Trasera , Proteínas de Andamiaje Homer/metabolismo , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Prueba de Campo Abierto , Prosencéfalo , Receptor de Adenosina A1/metabolismo , Recompensa , Privación de Sueño/psicología
7.
Neuron ; 104(2): 338-352.e7, 2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31420117

RESUMEN

Conventional antidepressants have limited efficacy and many side effects, highlighting the need for fast-acting and specific medications. Induction of the synaptic protein Homer1a mediates the effects of different antidepressant treatments, including the rapid action of ketamine and sleep deprivation (SD). We show here that mimicking Homer1a upregulation via intravenous injection of cell-membrane-permeable TAT-Homer1a elicits rapid antidepressant effects in various tests. Similar to ketamine and SD, in vitro and in vivo application of TAT-Homer1a enhances mGlu5 signaling, resulting in increased mTOR pathway phosphorylation, and upregulates synaptic AMPA receptor expression and activity. The antidepressant action of SD and Homer1a induction depends on mGlu5 activation specifically in excitatory CaMK2a neurons and requires enhanced AMPA receptor activity, translation, and trafficking. Moreover, our data demonstrate a pronounced therapeutic potential of different TAT-fused peptides that directly modulate mGlu5 and AMPA receptor activity and thus might provide a novel strategy for rapid and effective antidepressant treatment.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Trastorno Depresivo Mayor/metabolismo , Proteínas de Andamiaje Homer/farmacología , Receptor del Glutamato Metabotropico 5/efectos de los fármacos , Receptores AMPA/efectos de los fármacos , Sinapsis/efectos de los fármacos , Animales , Trastorno Depresivo Mayor/genética , Modelos Animales de Enfermedad , Productos del Gen tat , Proteínas de Andamiaje Homer/genética , Proteínas de Andamiaje Homer/metabolismo , Ratones , Ratones Noqueados , Fragmentos de Péptidos , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores AMPA/metabolismo , Transducción de Señal/efectos de los fármacos , Privación de Sueño/metabolismo , Sinapsis/metabolismo , Serina-Treonina Quinasas TOR/efectos de los fármacos , Regulación hacia Arriba
8.
J Neurochem ; 105(3): 904-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18088370

RESUMEN

Both adenosine and interleukin-6 (IL-6) have been implicated in the pathophysiology of, e.g., epileptic seizures, traumatic brain injury, and affective disorders. Stimulation of adenosine A2B receptors on astrocytes in vitro leads to the increased synthesis and secretion of IL-6. We investigated whether or not activation of adenosine receptors evokes an increase of IL-6 release also in vivo. 5'-N-ethylcarboxamidoadenosine, a non-specific adenosine-agonist or vehicle was administered into the striatum of freely moving mice by reverse microdialysis. A statistical significant increase of the IL-6 concentration in the perfusate was detected already 60 min after 5'-N-ethylcarboxamidoadenosine administration. IL-6 increased progressively and reached a maximum after 240 min. This effect appears to be mediated through adenosine A2B receptors since it was counteracted by the specific A2B receptor antagonist MRS1706 but not by the specific A1 receptor antagonist DPCPX. We conclude that adenosine via activation of A2B receptors evokes IL-6 release also in vivo.


Asunto(s)
Adenosina/metabolismo , Cuerpo Estriado/metabolismo , Interleucina-6/metabolismo , Receptor de Adenosina A2B/metabolismo , Adenosina/agonistas , Agonistas del Receptor de Adenosina A2 , Antagonistas del Receptor de Adenosina A2 , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Química Encefálica/efectos de los fármacos , Química Encefálica/fisiología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/inmunología , Encefalitis/inmunología , Encefalitis/metabolismo , Encefalitis/fisiopatología , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Interleucina-6/inmunología , Ratones , Ratones Endogámicos BALB C , Microdiálisis , Purinas/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Vasodilatadores/farmacología
9.
J Affect Disord ; 109(1-2): 65-73, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18067973

RESUMEN

BACKGROUND: Clinical guidelines recommend the combination of pharmaco- and psychotherapy for the treatment of chronic depression, although there are only a few studies supporting an additive effect of psychotherapy. METHODS: Forty-five inpatients with a chronic Major Depressive Disorder were randomized to 5 weeks of either Interpersonal Psychotherapy (IPT) modified for an inpatient setting (15 individual and 8 group sessions) plus pharmacotherapy or to medication plus Clinical Management (CM). The 17-item Hamilton Rating Scale for Depression was the primary outcome measure. The study included a prospective naturalistic follow-up, 3- and 12-months after discharge. RESULTS: Intent-to-treat analyses revealed a significantly greater reduction of depressive symptoms as well as better global functioning of patients treated with IPT compared to the CM group at week 5. Response and sustained response rates differed significantly between the two treatment conditions, favouring the IPT group. Remission rates were considerably higher for IPT patients who completed the treatment (67% vs. 32%). Patients who initially responded to IPT exhibited greater treatment gains at 12 months since only 7% of these subjects relapsed compared with 25% of the CM subjects. In the long-term, additional IPT led to a lower symptom level and higher global functioning. LIMITATIONS: The study uses data of a subset of patients from a larger trial. Both treatment groups did not receive comparable amounts of therapeutic attention. Extrapolating the data from this inpatient study to chronically depressed outpatients may not be possible. CONCLUSIONS: Intensive combined treatment provides superior acute and long-term effects over standard treatment in chronically depressed inpatients.


Asunto(s)
Amitriptilina/uso terapéutico , Antidepresivos/uso terapéutico , Trastorno Depresivo Mayor/terapia , Psicoterapia de Grupo/métodos , Sertralina/uso terapéutico , Adolescente , Adulto , Anciano , Enfermedad Crónica , Terapia Combinada , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/rehabilitación , Esquema de Medicación , Femenino , Estudios de Seguimiento , Hospitalización , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Resultado del Tratamiento
10.
Neurosci Biobehav Rev ; 88: 63-72, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29550211

RESUMEN

There is an urgent, unmet clinical need for faster and more efficient antidepressant drugs with higher response rates. In animal models of depression it was shown in the last few years that inhibition of three signaling molecules (BDNF, p11 and Homer1a) prevents efficacy of antidepressant therapy. These data not only show the crucial role of these factors for the treatment of depression, but may also point towards a better understanding of the molecular changes responsible for successful antidepressant therapy. Reviewing the literature concerning BNDF, p11 and Homer1a we here describe a molecular network in which these molecules interact with each other finally leading to facilitation of AMPA receptor signaling and plasticity, corroborating the current idea of AMPA receptors being a promising drug target in depression.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Trastorno Depresivo/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Humanos , Transducción de Señal/fisiología , Resultado del Tratamiento
11.
Am J Psychiatry ; 164(5): 768-77, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17475736

RESUMEN

OBJECTIVE: The purpose of this article was to determine the relative efficacy of a psychotherapy program when combined with pharmacotherapy versus medication and clinical management in more severely depressed patients. METHOD: A randomized controlled trial was conducted in 124 hospitalized patients with DSM-IV major depressive disorder that compared 5 weeks of interpersonal psychotherapy modified for depressed inpatients (15 individual and eight group sessions) plus pharmacotherapy with a regimen that involved medication plus intensive clinical management. The study included a prospective, naturalistic follow-up 3 and 12 months after acute treatment in 97 of 105 treatment completers. The 17-item version of the Hamilton Depression Rating Scale (HAM-D) was the primary outcome measure. RESULTS: For the intent-to-treat cohort (N=124), analysis of covariance (ANCOVA) showed that patients treated with interpersonal psychotherapy had a significantly greater reduction of depressive symptoms at week 5. Response rates differed significantly between the two treatment conditions, favoring the group that received adjuvant interpersonal psychotherapy (70%) versus clinical management (51%). Remission rates also tended to be higher for patients in the interpersonal psychotherapy group (49% versus 34%). Patients who initially responded to interpersonal psychotherapy exhibited greater treatment gains at the 3-month follow-up evaluation, since only 3% of these subjects relapsed, compared with 25% of the clinical management subjects. Nine months later, this difference lost statistical significance. CONCLUSIONS: An inpatient treatment program with both brief and intensive psychotherapy plus pharmacotherapy is superior to standard treatment. The results, which add to a growing body of evidence, suggest that this combination treatment may offer an advantage over treatment with medication and clinical management for more severely depressed patients.


Asunto(s)
Antidepresivos/uso terapéutico , Trastorno Depresivo Mayor/terapia , Hospitalización , Psicoterapia/métodos , Adulto , Amitriptilina/uso terapéutico , Antidepresivos Tricíclicos/uso terapéutico , Estudios de Cohortes , Terapia Combinada , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/psicología , Femenino , Estudios de Seguimiento , Humanos , Relaciones Interpersonales , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Escalas de Valoración Psiquiátrica/estadística & datos numéricos , Psicoterapia de Grupo , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Sertralina/uso terapéutico , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
12.
J Affect Disord ; 91(2-3): 195-203, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16488022

RESUMEN

BACKGROUND: The aim of the study was to identify risk factors in subjects at risk for depressive disorders and controls. METHODS: In a 6.5 year follow-up study we examined the effects of personality (neuroticism, frustration intolerance, rigidity, melancholic type), adverse life events and chronic difficulties on depressive symptoms in 89 high-risk subjects (HRS, siblings and children of patients suffering from an affective disorder), without any mental illness at wave 1 (T1), and 49 controls without any personal and family history of psychiatric disorder at T1. To this end, regression analysis and path analysis using a structural equation model (only for HRS) were performed. RESULTS: Risk factors for depressive symptoms at wave 2 (T2) in HRS comprised acute adverse life events, frustration intolerance (T1) and depressive symptoms (T1). Risk factors for depressive symptoms in controls included chronic difficulties, neuroticism and rigidity. HRS had less stressful life events and the same risk for chronic difficulties, but perceived adverse events as more stressful. LIMITATION: The sample size of the control group is too small for identifying slight effects. CONCLUSION: Our results indicate that the impact on the emergence of depressive symptoms of various risk factors is different in high-risk subjects and controls. High-risk subjects are more sensitive to the depressogenic effects of acute stress and thus avoid potential stressful changes in their life to a higher extent. On the other hand, the influence of persistent factors such as personality traits (neuroticism, rigidity) and chronic difficulties on subsequent depressive symptoms was less pronounced in HRS as compared to controls.


Asunto(s)
Depresión/epidemiología , Trastornos del Humor/genética , Personalidad , Estrés Psicológico/epidemiología , Adulto , Depresión/diagnóstico , Depresión/psicología , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Humanos , Masculino , Trastornos del Humor/diagnóstico , Trastornos del Humor/psicología , Factores de Riesgo , Estrés Psicológico/psicología , Encuestas y Cuestionarios
13.
Front Psychiatry ; 7: 194, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28008318

RESUMEN

Bipolar disorder (BD) is a severe and lifelong condition. Primary endogenic polygenetic forms are common. Secondary organic forms have received increasing interest recently due to the detection of immunological encephalopathies that mimic various psychiatric syndromes, including BD. However, only limited data about routine findings of cerebrospinal fluid (CSF) analyses in BD are available. Therefore, we investigated the frequency of alterations in the CSF in patients with BD and the association with autoantibodies, cerebral magnetic resonance imaging, and electroencephalography findings. CSF samples of patients with BD collected from January 1998 until December 2015 were analyzed retrospectively. Patients with preexisting causes for alterations in the CSF (e.g., patients with obvious past or current neurological disorders) were excluded. In total, 63 patients with BD fulfilled the inclusion criteria for the study. In 1.6% of the patients with BD, an increased white blood cell count was found in the CSF. Increased albumin quotients were found in 12.9% of the patients, oligoclonal bands (OCBs) in 1.6%, and increased immunoglobulin (Ig) G indices in 3.2% (OCBs were not measured in case of increased IgG indices). No significant differences in CSF findings were found between patients with manic and depressive episodes. The main findings of this open uncontrolled study are that alterations in the CSF may be found in a small, but potentially relevant, subgroup of patients with BD. These findings are discussed in light of the new concepts of mild encephalitis and immunological encephalopathy. The detection of patients with possibly secondary organic bipolar syndromes could open up new causal treatment options with immunomodulatory medication.

14.
Sleep Med Rev ; 30: 53-62, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26803484

RESUMEN

Therapeutic sleep deprivation (SD) is a rapid acting treatment for major depressive disorder (MDD). Within hours, SD leads to a dramatic decrease in depressive symptoms in 50-60% of patients with MDD. Scientifically, therapeutic SD presents a unique paradigm to study the neurobiology of MDD. Yet, up to now, the neurobiological basis of the antidepressant effect, which is most likely different from today's first-line treatments, is not sufficiently understood. This article puts the idea forward that sleep/wake-dependent shifts in synaptic plasticity, i.e., the neural basis of adaptive network function and behavior, represent a critical mechanism of therapeutic SD in MDD. Particularly, this article centers on two major hypotheses of MDD and sleep, the synaptic plasticity hypothesis of MDD and the synaptic homeostasis hypothesis of sleep-wake regulation, and on how they can be integrated into a novel synaptic plasticity model of therapeutic SD in MDD. As a major component, the model proposes that therapeutic SD, by homeostatically enhancing cortical synaptic strength, shifts the initially deficient inducibility of associative synaptic long-term potentiation (LTP) in patients with MDD in a more favorable window of associative plasticity. Research on the molecular effects of SD in animals and humans, including observations in the neurotrophic, adenosinergic, monoaminergic, and glutamatergic system, provides some support for the hypothesis of associative synaptic plasticity facilitation after therapeutic SD in MDD. The model proposes a novel framework for a mechanism of action of therapeutic SD that can be further tested in humans based on non-invasive indices and in animals based on direct studies of synaptic plasticity. Further determining the mechanisms of action of SD might contribute to the development of novel fast acting treatments for MDD, one of the major health problems worldwide.


Asunto(s)
Trastorno Depresivo Mayor/fisiopatología , Trastorno Depresivo Mayor/terapia , Plasticidad Neuronal , Privación de Sueño/fisiopatología , Animales , Trastorno Depresivo Mayor/psicología , Humanos , Sueño/fisiología , Vigilia/fisiología
15.
Eur Neuropsychopharmacol ; 26(4): 705-16, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26899588

RESUMEN

Patients with Major Depressive Disorder (MDD) and no improvement after two weeks of antidepressant pharmacotherapy have a high risk of treatment failure. The aim of the study was to determine whether an early medication change (EMC) strategy is superior to a guideline-based treatment in MDD patients without improvement after two weeks of antidepressant pharmacotherapy. Eight-hundred-and-eighty-nine patients with MDD were enrolled, 879 patients received the SSRI escitalopram. Of those, 192 patients had no improvement, defined as a reduction of < 20% on the Hamilton Depression Rating Scale (HAMD-17) after 14 days of treatment, and were randomly assigned to open treatment with the EMC strategy (n = 97; venlafaxine XR for study days 15-56; in case of sustained non-improvement on day 28, lithium augmentation for days 29-56) or TAU (n = 95; escitalopram continuation; non-responders on day 28 were switched to venlafaxine XR for four weeks, i.e. days 29-56). The primary outcome was remission (HAMD-17 ≤ 7) after 8 weeks of treatment as assessed by blinded raters. Remission rates were 24% for EMC and 16% for TAU, which was not significantly different (p = 0.2056). Sensitivity analyses for the primary and secondary effectiveness endpoints consistently showed favorable results for patients randomized to EMC. The results confirm data from post-hoc analyses of clinical trials showing that early non-improvement identifies patients who likely need alternate interventions. However, the herein used two-step switch/augmentation strategy for this risk group was not more effective than the control intervention. Alternate strategies and other design aspects are discussed in order to support researchers addressing the same research question.


Asunto(s)
Antidepresivos/administración & dosificación , Antidepresivos/uso terapéutico , Citalopram/uso terapéutico , Trastorno Depresivo Resistente al Tratamiento/tratamiento farmacológico , Intervención Médica Temprana , Clorhidrato de Venlafaxina/uso terapéutico , Adolescente , Adulto , Anciano , Antidepresivos/efectos adversos , Antidepresivos de Segunda Generación/uso terapéutico , Citalopram/administración & dosificación , Citalopram/efectos adversos , Preparaciones de Acción Retardada/uso terapéutico , Quimioterapia Combinada , Femenino , Humanos , Litio/uso terapéutico , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Clorhidrato de Venlafaxina/administración & dosificación , Clorhidrato de Venlafaxina/efectos adversos , Adulto Joven
16.
Neurochem Int ; 46(6): 501-12, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15769552

RESUMEN

Adenosine binds to a class of G-protein coupled receptors, which are further distinguished as A(1), A(2a), A(2b) and A(3) adenosine receptors. As we have shown earlier, the stable adenosine analogue NECA (N6-(R)-phenylisopropyladenosine) stimulates IL-6 expression in the human astrocytoma cell line U373 MG via the A(2b) receptor. The mechanism by which NECA promotes astrocytic IL-6 expression has not been identified. By using various inhibitors of signal transduction, we found that p38 mitogen-activated protein kinases (MAPK) activation (inhibitor SB202190), but not extracellular signal-regulated kinase (ERK) (PD98059) and c-jun N-terminal kinase (JNK)(SP600125), is essential in the NECA-induced signalling cascade that leads to the increase in IL-6 synthesis in U373 MG cells. Results obtained with protein kinase C (PKC) inhibitors that have different substrate specificities, indicated that the PKC delta and epsilon isoforms are also involved in adenosine receptor A(2b) dependent upregulation of IL-6 expression. This is supported by the fact that NECA induced the activation of PKC delta and epsilon in U373 MG cells.


Asunto(s)
Adenosina/metabolismo , Astrocitoma/metabolismo , Interleucina-6/metabolismo , Proteína Quinasa C/metabolismo , Receptor de Adenosina A2B/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Agonistas del Receptor de Adenosina A2 , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C-delta , Proteína Quinasa C-epsilon , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
17.
J Affect Disord ; 84(1): 33-42, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15620383

RESUMEN

BACKGROUND: Particular patterns of personality (e.g., neuroticism, obsessionality) and difficulties in various social roles have been found to be associated with unipolar depression. Interpersonal and instrumental difficulties of depressives can be understood either as a risk factor, or as a consequence caused by the disorder itself. Concerning patients with bipolar disorder, there is some evidence that their premorbid level of occupational and educational achievement is often superior when compared to the premorbid functioning of patients with unipolar depression. METHODS: Personality features and the level of social functioning of 114 high-risk subjects (healthy first-degree relatives of patients suffering from an affective disorder) have been investigated using self- and expert-ratings. Sixty-three subjects without a personal and family history of psychiatric disorder served as the reference group. RESULTS: Relatives of melancholic depressives described themselves as more neurotic than controls but proved to be inconspicuous regarding their role functioning. Relatives of bipolar I patients were more strongly oriented toward social norms, and their instrumental role functioning was superior to that of controls. Neuroticism was strongly associated with depressive symptoms. LIMITATION: The statistical power of our data is sufficient to detect medium effect sizes but is insufficient for identifying small group differences. CONCLUSION: Whether these discriminating personality features and other variables (not characterising the high-risk group (HRG) as a whole) act as true vulnerability factor have to be clarified by a follow-up investigation.


Asunto(s)
Trastornos del Humor/epidemiología , Trastornos del Humor/genética , Trastornos de la Personalidad/epidemiología , Conducta Social , Adolescente , Adulto , Manual Diagnóstico y Estadístico de los Trastornos Mentales , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Trastornos del Humor/diagnóstico , Trastornos de la Personalidad/diagnóstico , Factores de Riesgo , Índice de Severidad de la Enfermedad
18.
PLoS One ; 10(8): e0134934, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26263491

RESUMEN

The "regulators of g-protein signalling" (RGS) comprise a large family of proteins that limit by virtue of their GTPase accelerating protein domain the signal transduction of G-protein coupled receptors. RGS proteins have been implicated in various neuropsychiatric diseases such as schizophrenia, drug abuse, depression and anxiety and aggressive behaviour. Since conditions associated with a large increase of adenosine in the brain such as seizures or ischemia were reported to modify the expression of some RGS proteins we hypothesized that adenosine might regulate RGS expression in neural cells. We measured the expression of RGS-2,-3, and -4 in both transformed glia cells (human U373 MG astrocytoma cells) and in primary rat astrocyte cultures stimulated with adenosine agonists. Expression of RGS-2 mRNA as well as RGS2 protein was increased up to 30-fold by adenosine agonists in astrocytes. The order of potency of agonists and the blockade by the adenosine A2B-antagonist MRS1706 indicated that this effect was largely mediated by adenosine A2B receptors. However, a smaller effect was observed due to activation of adenosine A2A receptors. In astrocytoma cells adenosine agonists elicited an increase in RGS-2 expression solely mediated by A2B receptors. Expression of RGS-3 was inhibited by adenosine agonists in both astrocytoma cells and astrocytes. However while this effect was mediated by A2B receptors in astrocytoma cells it was mediated by A2A receptors in astrocytes as assessed by the order of potency of agonists and selective blockade by the specific antagonists MRS1706 and ZM241385 respectively. RGS-4 expression was inhibited in astrocytoma cells but enhanced in astrocytes by adenosine agonists.


Asunto(s)
Astrocitos/metabolismo , Reguladores de Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Receptores de Adenosina A2/metabolismo , Adenosina/farmacología , Agonistas del Receptor de Adenosina A2/farmacología , Animales , Astrocitoma/genética , Astrocitoma/metabolismo , Línea Celular Tumoral , Humanos , Proteínas RGS/genética , ARN Mensajero/genética , Ratas
19.
Neuron ; 87(3): 549-62, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26247862

RESUMEN

Major depressive disorder is among the most commonly diagnosed disabling mental diseases. Several non-pharmacological treatments of depression upregulate adenosine concentration and/or adenosine A1 receptors (A1R) in the brain. To test whether enhanced A1R signaling mediates antidepressant effects, we generated a transgenic mouse with enhanced doxycycline-regulated A1R expression, specifically in forebrain neurons. Upregulating A1R led to pronounced acute and chronic resilience toward depressive-like behavior in various tests. Conversely, A1R knockout mice displayed an increased depressive-like behavior and were resistant to the antidepressant effects of sleep deprivation (SD). Various antidepressant treatments increase homer1a expression in medial prefrontal cortex (mPFC). Specific siRNA knockdown of homer1a in mPFC enhanced depressive-like behavior and prevented the antidepressant effects of A1R upregulation, SD, imipramine, and ketamine treatment. In contrast, viral overexpression of homer1a in the mPFC had antidepressant effects. Thus, increased expression of homer1a is a final common pathway mediating the antidepressant effects of different antidepressant treatments.


Asunto(s)
Proteínas Portadoras/biosíntesis , Depresión/metabolismo , Imipramina/uso terapéutico , Ketamina/uso terapéutico , Receptor de Adenosina A1/biosíntesis , Privación de Sueño/metabolismo , Animales , Depresión/psicología , Depresión/terapia , Proteínas de Andamiaje Homer , Humanos , Imipramina/farmacología , Ketamina/farmacología , Ratones , Ratones Noqueados , Ratones Transgénicos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Receptor de Adenosina A1/deficiencia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Privación de Sueño/psicología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA