Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Brain Behav Immun ; 80: 763-776, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31108171

RESUMEN

Acute stressors can induce fear and physiologic responses that prepare the body to protect from danger. A key component of this response is immune system readiness. In particular, inflammasome activation appears critical to linking stress to the immune system. Here, we show that a novel combination of handling procedures used regularly in mouse research impairs novel object recognition (NOR) and activates caspase-1 in the amygdala. In male mice, this handling-stress paradigm combined weighing, scruffing and sham abdominal injection once per hr. While one round of weigh/scruff/needle-stick had no impact on NOR, two rounds compromised NOR without impacting location memory or anxiety-like behaviors. Caspase-1 knockout (KO), IL-1 receptor 1 (IL-1R1) KO and IL-1 receptor antagonist (IL-RA)-administered mice were resistant to handling stress-induced loss of NOR. In addition, examination of the brain showed that handling stress increased caspase-1 activity 85% in the amygdala without impacting hippocampal caspase-1 activity. To delineate danger signals relevant to handling stress, caffeine-administered and adenosine 2A receptor (A2AR) KO mice were tested and found resistant to impaired learning and caspase-1 activation. Finally, mice treated with the ß-adrenergic receptor antagonist, propranolol, were resistant to handling stress-induced loss of NOR and caspase-1 activation. Taken together, these results indicate that handling stress-induced impairment of object learning is reliant on a pathway requiring A2AR-dependent activation of caspase-1 in the amygdala that appears contingent on ß-adrenergic receptor functionality.


Asunto(s)
Adenosina/metabolismo , Caspasa 1/metabolismo , Aprendizaje/fisiología , Estrés Psicológico/metabolismo , Adenosina/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad , Encéfalo/metabolismo , Activación Enzimática , Miedo/fisiología , Hipocampo/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Receptores Purinérgicos P1/metabolismo , Transducción de Señal/efectos de los fármacos
2.
Epilepsia ; 60(4): 626-635, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30770561

RESUMEN

OBJECTIVE: Viral encephalitis increases the risk for developing seizures and epilepsy. Indoleamine 2,3-dioxygenase 1 (Ido1) is induced by inflammatory cytokines and functions to metabolize tryptophan to kynurenine. Kynurenine can be further metabolized to produce kynurenic acid and the N-methyl-d-aspartate receptor agonist quinolinic acid (QuinA). In the present study, we sought to determine the role of Ido1 in promoting seizures in an animal model of viral encephalitis. METHODS: C57BL/6J and Ido1 knockout mice (Ido1-KO) were infected with Theiler's murine encephalomyelitis virus (TMEV). Quantitative real-time polymerase chain reaction was used to evaluate hippocampal expression of proinflammatory cytokines, Ido1, and viral RNA. Body weights and seizure scores were recorded daily. Elevated zero maze was used to assess differences in behavior, and hippocampal pathology was determined by immunohistochemistry. RESULTS: Infected C57BL/6J mice up-regulated proinflammatory cytokines, Ido1, and genes encoding the enzymatic cascade responsible for QuinA production in the kynurenine pathway prior to the onset of seizures. Seizure incidence was elevated in Ido1-KO compared to C57BL/6J mice. Infection increased locomotor activity in Ido1-KO compared to C57BL/6J mice. Furthermore, the occurrence of seizures was associated with hyperexcitability. Neither expression of proinflammatory cytokines nor viral RNA was altered as a result of genotype. Immunohistochemical analysis revealed increased hippocampal pathology in Ido1-KO mice. SIGNIFICANCE: Our findings suggest that Ido1 deletion promotes seizures and neuropathogenesis during acute TMEV encephalitis.


Asunto(s)
Encefalitis Viral/complicaciones , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Convulsiones/enzimología , Animales , Infecciones por Cardiovirus/complicaciones , Modelos Animales de Enfermedad , Encefalitis Viral/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Convulsiones/virología , Theilovirus
3.
Brain Behav Immun ; 69: 321-335, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29241670

RESUMEN

Behavioral symptoms associated with mood disorders have been intimately linked with immunological and psychological stress. Induction of immune and stress pathways is accompanied by increased tryptophan entry into the Kynurenine (Kyn) Pathway as governed by the rate-limiting enzymes indoleamine/tryptophan 2,3-dioxygenases (DO's: Ido1, Ido2, Tdo2). Indeed, elevated DO expression is associated with inflammation- and stress-related depression symptoms. Here we examined central (brain, astrocyte and microglia) and peripheral (lung, liver and spleen) DO expression in mice treated intraperitoneally with lipopolysaccharide (LPS) and dexamethasone (DEX) to model the response of the Kyn Pathway to inflammation and glucocorticoids. LPS-induced expression of cytokines in peripheral tissues was attenuated by DEX, confirming inflammatory and anti-inflammatory responses, respectively. Increased Kyn levels following LPS and DEX administration verified Kyn Pathway activation. Expression of multiple mRNA isoforms for each DO, which we have shown to be differentially utilized and regulated, were quantified including reference/full-length (FL) and variant (v) transcripts. LPS increased Ido1-FL in brain (∼1000-fold), a response paralleled by increased expression in both astrocytes and microglia. Central Ido1-FL was not changed by DEX; however, LPS-induced Ido1-FL was decreased by DEX in peripheral tissues. In contrast, DEX increased Ido1-v1 expression by astrocytes and microglia, but not peripheral tissues. In comparison, brain Ido2 was minimally induced by LPS or DEX. Uniquely, Ido2-v6 was LPS- and DEX-inducible in astrocytes, suggesting a unique role for astrocytes in response to inflammation and glucocorticoids. Only DEX increased central Tdo2 expression; however, peripheral Tdo2 was upregulated by either LPS or DEX. In summary, specific DO isoforms are increased by LPS and DEX, but LPS-dependent Ido1 and Ido2 induction are attenuated by DEX only in the periphery indicating that elevated DO expression and Kyn production within the brain can occur independent of the periphery. These findings demonstrate a plausible interaction between immune activation and glucocorticoids associated with depression.


Asunto(s)
Dexametasona/farmacología , Glucocorticoides/farmacología , Quinurenina/metabolismo , Lipopolisacáridos/farmacología , Neuroglía/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Citocinas/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Neuroglía/metabolismo , Estrés Psicológico/metabolismo
4.
Semin Immunol ; 26(5): 389-93, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24556600

RESUMEN

Twenty-five years ago, immunologists and neuroscientists had little science of mutual interest. This is no longer the case. Neuroscientists now know that the first formally defined cytokine, IL-1, activates a discrete population of hypothalamic neurons. This interaction leads to the release of glucocorticoids from the adrenal gland, a hormone that has a long history in immunoregulation. Immunologists have been surprised to learn that lymphoid cells synthesize acetylcholine, the first formally recognized neurotransmitter. This neurotransmitter suppresses the synthesis of TNF. These discoveries blur the distinction of neuroscience and immunology as distinct disciplines. There are now 37 formally recognized cytokines and their receptors, and at least 60 classical neurotransmitters plus over 50 neuroactive peptides. These findings explain why both immunologists and neuroscientists are getting nervous about immunity and highlight a real need to apply integrative physiological approaches in biomedical research.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Hipotálamo/metabolismo , Sistema Inmunológico/metabolismo , Linfocitos/metabolismo , Neuronas/metabolismo , Sistemas Neurosecretores/metabolismo , Acetilcolina/inmunología , Acetilcolina/metabolismo , Glándulas Suprarrenales/inmunología , Animales , Citocinas/inmunología , Citocinas/metabolismo , Glucocorticoides/inmunología , Glucocorticoides/metabolismo , Humanos , Hipotálamo/inmunología , Linfocitos/inmunología , Neuronas/inmunología , Sistemas Neurosecretores/inmunología , Neurotransmisores/inmunología , Neurotransmisores/metabolismo , Transducción de Señal
5.
Brain Behav Immun ; 62: 24-29, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28179106

RESUMEN

Glioblastoma (GBM) is the most common malignant brain tumor in adults with a median survival of 14.6months. A contributing factor to GBM aggressiveness is the intratumoral expression of the potently immunosuppressive enzyme, indoleamine 2,3 dioxygenase 1 (IDO1). The enzymatic activity of IDO1 is associated with the conversion of tryptophan into downstream kynurenine (Kyn), which has previously been hypothesized to contribute toward the suppression of tumor immunity. Utilizing the syngeneic, immunocompetent, intracranial GL261 cell GBM model, we previously demonstrated that tumor cell, but not non-tumor cell IDO1, suppresses T cell-mediated brain tumor regression in mice. Paradoxically, we also showed that the survival advantage mediated by immune checkpoint blockade is abrogated by non-tumor cell IDO1 deficiency. Here, we have built on our past observations and confirm the maladaptive role of tumor cell IDO1 in a novel mouse GBM model. We also demonstrate that, non-tumor cells, rather than mouse GBM cells, are the dominant contributor to IDO1-mediated enzyme activity. Finally, we show the novel associations between maximally-effective immune-checkpoint blockade-mediated survival, non-tumor cell IDO1 and intra-GBM Kyn levels. These data suggest for the first time that, GBM cell-mediated immunosuppression is IDO1 enzyme independent, while the survival benefits of immune checkpoint blockade require non-tumor cell IDO1 enzyme activity. Given that current clinical inhibitors vary in their mechanism of action, in terms of targeting IDO1 enzyme activity versus enzyme-independent effects, this work suggests that choosing an appropriate IDO1 pharmacologic will maximize the effectiveness of future immune checkpoint blockade approaches.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Glioblastoma/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Quinurenina/metabolismo , Animales , Encéfalo/patología , Neoplasias Encefálicas/patología , Modelos Animales de Enfermedad , Glioblastoma/patología , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Ratones , Ratones Noqueados
6.
Brain Behav Immun ; 62: 219-229, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28212884

RESUMEN

Abundant evidence connects depression symptomology with immune system activation, stress and subsequently elevated levels of kynurenine. Anti-depressants, such as the tricyclic norepinephrine/serotonin reuptake inhibitor desipramine (Desip), were developed under the premise that increasing extracellular neurotransmitter level was the sole mechanism by which they alleviate depressive symptomologies. However, evidence suggests that anti-depressants have additional actions that contribute to their therapeutic potential. The Kynurenine Pathway produces tryptophan metabolites that modulate neurotransmitter activity. This recognition identified another putative pathway for anti-depressant targeting. Considering a recognized role of the Kynurenine Pathway in depression, we investigated the potential for Desip to alter expression of rate-limiting enzymes of this pathway: indoleamine-2,3-dioxygenases (Ido1 and Ido2). Mice were administered lipopolysaccharide (LPS) or synthetic glucocorticoid dexamethasone (Dex) with Desip to determine if Desip alters indoleamine-dioxygenase (DO) expression in vivo following a modeled immune and stress response. This work was followed by treating murine and human peripheral blood mononuclear cells (PBMCs) with interferon-gamma (IFNγ) and Desip. In vivo: Desip blocked LPS-induced Ido1 expression in hippocampi, astrocytes, microglia and PBMCs and Ido2 expression by PBMCs. Ex vivo: Desip decreased IFNγ-induced Ido1 and Ido2 expression in murine PBMCs. This effect was directly translatable to the human system as Desip decreased IDO1 and IDO2 expression by human PBMCs. These data demonstrate for the first time that an anti-depressant alters expression of Ido1 and Ido2, identifying a possible new mechanism behind anti-depressant activity. Furthermore, we propose the assessment of PBMCs for anti-depressant responsiveness using IDO expression as a biomarker.


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Desipramina/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Animales , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Interferón gamma/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Adulto Joven
7.
J Neuroinflammation ; 13(1): 98, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27142940

RESUMEN

BACKGROUND: Increased tryptophan metabolism towards the production of kynurenine via indoleamine/tryptophan-2,3-dioxygenases (DOs: Ido1, Ido2, and Tdo2) is strongly associated with the prevalence of major depressive disorder in patients and the induction of depression-like behaviors in animal models. Several studies have suggested that activation of the immune system or elevated corticosteroids drive DO expression; however, mechanisms linking cytokines, corticosteroids, and DOs to psychiatric diseases remain unclear. Various attempts have been made to correlate DO gene expression within the brain to behavior, but disparate results have been obtained. We believe that discrepancies arise as a result of the under-recognized existence of multiple mRNA transcripts for each DO. Unfortunately, there are no reports regarding how the multiple transcripts are distributed or regulated. Here, we used organotypic hippocampal slice cultures (OHSCs) to directly test the ability of inflammatory and stress mediators to differentially regulate DO transcripts. METHODS: OHSCs were treated with pro-inflammatory mediators (interferon-gamma (IFNγ), lipopolysaccharide (LPS), and polyinosine-polycytidylic acid (pI:C)) with or without corticosteroids (dexamethasone (Dex: glucocorticoid receptor (GR) agonist), aldosterone (Aldo: mineralocorticoid receptor (MR) agonist), or corticosterone (Cort: GR/MR agonist)). RESULTS: IFNγ induced Ido1-full length (FL) and Ido1-variant (v) expression, and surprisingly, Dex, Cort, and Aldo interacted with IFNγ to further elevate expression of Ido1, importantly, in a transcript dependent manner. IFNγ, LPS, and pI:C increased expression of Ido2-v1 and Ido2-v3 transcripts, whereas only IFNγ increased expression of Ido2-v2. Overall Ido2 transcripts were relatively unaffected by GR or MR activation. Naïve mouse brain expresses multiple Tdo2 transcripts. Dex and Cort induced expression of only one of the three Tdo2 transcripts (Tdo2-FL) in OHSCs. CONCLUSIONS: These results establish that multiple transcripts for all three DOs are expressed within the mouse hippocampus, under the control of distinct regulatory pathways. These data identify a previously unrecognized interaction between corticosteroid receptor activation and inflammatory signals on DO gene expression, which suggest that corticosteroids act to differentially enhance gene expression of Ido1, Ido2, and Tdo2.


Asunto(s)
Corticoesteroides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Quinurenina/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Hipocampo/efectos de los fármacos , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Reacción en Cadena de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
8.
Brain Behav Immun ; 44: 176-86, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25300921

RESUMEN

Behavioral indicators in the murine Bacille Calmette Guérin (BCG) model of inflammation have been studied individually; however, the variability of the behaviors across BCG levels and the mouse-to-mouse variation within BCG-treatment group are only partially understood. The objectives of this study were: (1) to gain a comprehensive understanding of sickness and depression-like behaviors in a BCG model of inflammation using multivariate approaches, and (2) to explore behavioral differences between BCG-treatment groups and among mice within group. Adult mice were challenged with either 0mg (saline), 5mg or 10mg of BCG (BCG-treatment groups: BCG0, BCG5, or BCG10, respectively) at Day 0 of the experiment. Sickness indicators included body weight changes between Day 0 and Day 2 and between Day 2 and Day 5, and horizontal locomotor activity and vertical activity (rearing) measured at Day 6. Depression-like indicators included duration of immobility in the forced swim test and in the tail suspension test at Day 6 and sucrose consumption in the sucrose preference test at Day 7. The simultaneous consideration of complementary sickness and depression-like indicators enabled a more precise characterization of behavioral changes associated with BCG-treatment and of mouse-to-mouse variation, relative to the analysis of indicators individually. Univariate and multivariate analyses confirmed differences between BCG-treatment groups in weight change early on the trial. Significant differences between BCG-treatment groups in depression-like behaviors were still measurable after Day 5. The potential for multivariate models to account for the correlation between behavioral indicators and to augment the analytical precision relative to univariate models was demonstrated both for sickness and for depression-like indicators. Unsupervised learning approaches revealed the complementary information provided by the sickness and depression-like indicators considered. Supervised learning approaches using cross-validation confirmed subtle differences between BCG-treatment groups and among mice within group identified by the consideration of sickness and depression-like indicators. These findings support the recommendation for multivariate and multidimensional analyses of sickness and depression-like indicators to augment the systemic understanding of the behavioral changes associated with infection.


Asunto(s)
Depresión/psicología , Conducta de Enfermedad , Infecciones por Mycobacterium/psicología , Animales , Conducta Animal , Peso Corporal , Depresión/etiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Análisis Multivariante , Infecciones por Mycobacterium/complicaciones , Mycobacterium bovis
9.
Brain Behav Immun ; 41: 218-31, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24907587

RESUMEN

Anxiety is one of the most commonly reported psychiatric conditions, but its pathogenesis is poorly understood. Ailments associated with activation of the innate immune system, however, are increasingly linked to anxiety disorders. In adult male mice, we found that adenosine doubled caspase-1 activity in brain by a pathway reliant on ATP-sensitive potassium (KATP) channels, protein kinase A (PKA) and the A2A adenosine receptor (AR). In addition, adenosine-dependent activation of caspase-1 increased interleukin (IL)-1ß in the brain by 2-fold. Peripheral administration of adenosine in wild-type (WT) mice led to a 2.3-fold increase in caspase-1 activity in the amygdala and to a 33% and 42% reduction in spontaneous locomotor activity and food intake, respectively, that were not observed in caspase-1 knockout (KO), IL-1 receptor type 1 (IL-1R1) KO and A2A AR KO mice or in mice administered a caspase-1 inhibitor centrally. Finally, adenosine administration increased anxiety-like behaviors in WT mice by 28% in the open field test and by 55% in the elevated zero-maze. Caspase-1 KO mice, IL-1R1 KO mice, A2A AR KO mice and WT mice treated with the KATP channel blocker, glyburide, were resistant to adenosine-induced anxiety-like behaviors. Thus, our results indicate that adenosine can act as an anxiogenic by activating caspase-1 and increasing IL-1ß in the brain.


Asunto(s)
Adenosina/toxicidad , Ansiedad/inducido químicamente , Encéfalo/metabolismo , Caspasa 1/fisiología , Interleucina-1beta/biosíntesis , Proteínas del Tejido Nervioso/fisiología , Receptor de Adenosina A2A/fisiología , Adenosina/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/fisiopatología , Carbazoles/farmacología , Caspasa 1/deficiencia , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Activación Enzimática/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Gliburida/farmacología , Interleucina-1beta/genética , Interleucina-1beta/fisiología , Transporte Iónico/efectos de los fármacos , Canales KATP/fisiología , Locomoción/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Potasio/metabolismo , Pirroles/farmacología , Receptor de Adenosina A2A/deficiencia , Receptor de Adenosina A2A/efectos de los fármacos , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/fisiología
10.
J Neuroinflammation ; 10: 54, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23634700

RESUMEN

BACKGROUND: Interleukin-1 beta converting enzyme (ICE, caspase 1) is a cysteine protease that processes immature pro-IL-1ß into active mature IL-1ß. IL-1ß is a pro-inflammatory cytokine that mediates many of the physiological and behavioral responses to inflammation. Genetic deletion of ICE has previously been shown to prevent some negative physiologic responses to lipopolysaccharide (LPS)-induced inflammation. METHODS: Here we used a preclinical murine model to test the hypothesis that ICE is necessary for development of depression-like behaviors following intracerebroventricular (ICV) treatment with LPS. Adult male ICE knockout (ICE KO) and congenic wild-type C57BL/6 J (WT) mice were administered LPS either ICV at 100 ng/mouse or intraperitoneally (IP) at 830 µg/kg body weight or an equal volume of saline as controls. Mice were monitored up to 48 h after treatment for both sickness and depression-like behaviors. RESULTS: LPS given ICV induced a loss of body weight in both WT and ICE KO mice. This sickness response was similar between WT and ICE KO mice. As expected, LPS administered ICV increased immobility in the forced swim test (FST) and decreased sucrose preference in WT mice but no change in either of these two depression-like behaviors was observed in ICE KO mice. Expression of TNF-α and CD11b in brain was lower in ICE-KO mice at 24 h following ICV administration of LPS compared to WT mice. In contrast, when LPS was given systemically, sickness response, depression-like behaviors, and expression of these genes were similar between the two strains of mice. CONCLUSIONS: These findings indicate that ICE plays a specific role in depression-like behavior induced by a central inflammatory stimuli even though it is not required when LPS is administered systemically.


Asunto(s)
Caspasa 1/metabolismo , Depresión/inducido químicamente , Depresión/enzimología , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Animales , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Química Encefálica/genética , Antígeno CD11b/biosíntesis , Caspasa 1/genética , Inhibidores de Caspasas/farmacología , Citocinas/metabolismo , Depresión/psicología , Preferencias Alimentarias , Inyecciones Intraventriculares , Lipopolisacáridos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Sacarosa , Natación/psicología , Factor de Necrosis Tumoral alfa/biosíntesis
11.
J Neuroinflammation ; 10: 87, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23866724

RESUMEN

BACKGROUND: Activation of the tryptophan degrading enzyme indoleamine-2,3-dioxygenase 1 (IDO1) is associated with the development of behavioral signs of depression. Systemic immune challenge induces IDO1 in both the periphery and the brain, leading to increased circulating and brain concentrations of kynurenines. However, whether IDO1 activity within the brain is necessary for the manifestation of depression-like behavior of mice following a central immune challenge remains to be elucidated. METHODS: We investigated the role of brain IDO1 in mediating depression-like behavior of mice in response to intracerebroventricular injection of saline or lipopolysaccharide (LPS, 10 ng). RESULTS: LPS increased the duration of immobility in the tail suspension test and decreased preference for a sucrose solution. These effects were associated with an activation of central but not peripheral IDO1, as LPS increased brain kynurenine but had no effect on plasma concentrations of kynurenine. Interestingly, genetic deletion or pharmacological inhibition of IDO1, using 1-methyl-tryptophan, abrogated the reduction in sucrose preference induced by intracerebroventricular LPS. 1-Methyl-tryptophan also blocked the LPS-induced increase in duration of immobility during the tail suspension test. CONCLUSIONS: These data indicate that activation of brain IDO1 is sufficient to induce depression-like behaviors of mice in response to central LPS.


Asunto(s)
Depresión/inducido químicamente , Depresión/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/deficiencia , Lipopolisacáridos/administración & dosificación , Animales , Antidepresivos/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Depresión/tratamiento farmacológico , Depresión/patología , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Preferencias Alimentarias/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Sacarosa/administración & dosificación , Factores de Tiempo , Triptófano/análogos & derivados , Triptófano/sangre , Triptófano/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos
12.
Brain Behav Immun ; 32: 63-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23454036

RESUMEN

Geriatric depression is a costly health issue, but little is known about its physiological underpinnings. Systemic inflammation sensitizes the innate immune system of aged animals and humans, but it is unknown if chronic, low-grade infections affect the duration of depressive-like behaviors. In this report, we infected adult (4-6 months) and aged (20-24 months) Balb/c mice with an attenuated strain of Mycobacterium bovis, Bacillus Calmette-Guérin (BCG), to induce a chronic infection. We then measured depression-like behaviors that have construct, face and predictive validity for human inflammation-associated clinical depression. Exposure to BCG caused acute sickness responses in both adult and aged mice. However, sickness behavior was prolonged in aged mice, as assessed by both locomotor and rearing activity. Two measures of depression-like behavior, which were tests involving sucrose preference and tail suspension, both showed that adult mice displayed depression-like behaviors at one day and seven days after exposure to BCG. However, aged mice continued to express both of these depression-like behaviors at three weeks following infection. Infection with BCG caused an increase in tryptophan catabolism, as evidenced by a significant rise in the plasma kynurenine/tryptophan ratio that peaked at 7 days post-infection. In aged mice, greater tryptophan catabolism persisted longer and remained elevated at 21 days post-infection. This finding is consistent with the prolonged duration of depression-like behaviors in aged mice. These are the first data using a chronic infection model to establish that recovery from inflammation-induced depression-like behavior and tryptophan catabolism are prolonged in aged animals.


Asunto(s)
Envejecimiento/psicología , Conducta Animal/fisiología , Depresión/psicología , Inflamación/psicología , Infecciones por Mycobacterium/psicología , Mycobacterium bovis , Anhedonia , Animales , Peso Corporal/fisiología , Enfermedad Crónica , Depresión/etiología , Depresión/metabolismo , Suspensión Trasera/psicología , Conducta de Enfermedad , Inflamación/etiología , Inflamación/metabolismo , Quinurenina/sangre , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Actividad Motora/fisiología , Infecciones por Mycobacterium/complicaciones , Infecciones por Mycobacterium/metabolismo , Bazo/patología , Triptófano/sangre
13.
J Exp Biol ; 216(Pt 1): 84-98, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23225871

RESUMEN

Humans and animals use the classical five senses of sight, sound, touch, smell and taste to monitor their environment. The very survival of feral animals depends on these sensory perception systems, which is a central theme in scholarly research on comparative aspects of anatomy and physiology. But how do all of us sense and respond to an infection? We cannot see, hear, feel, smell or taste bacterial and viral pathogens, but humans and animals alike are fully aware of symptoms of sickness that are caused by these microbes. Pain, fatigue, altered sleep pattern, anorexia and fever are common symptoms in both sick animals and humans. Many of these physiological changes represent adaptive responses that are considered to promote animal survival, and this constellation of events results in sickness behavior. Infectious agents display a variety of pathogen-associated molecular patterns (PAMPs) that are recognized by pattern recognition receptors (PRRs). These PRR are expressed on both the surface [e.g. Toll-like receptor (TLR)-4] and in the cytoplasm [e.g. nucleotide-binding oligomerization domain (Nod)-like receptors] of cells of the innate immune system, primarily macrophages and dendritic cells. These cells initiate and propagate an inflammatory response by stimulating the synthesis and release of a variety of cytokines. Once an infection has occurred in the periphery, both cytokines and bacterial toxins deliver this information to the brain using both humoral and neuronal routes of communication. For example, binding of PRR can lead to activation of the afferent vagus nerve, which communicates neuronal signals via the lower brain stem (nucleus tractus solitarius) to higher brain centers such as the hypothalamus and amygdala. Blood-borne cytokines initiate a cytokine response from vascular endothelial cells that form the blood-brain barrier (BBB). Cytokines can also reach the brain directly by leakage through the BBB via circumventricular organs or by being synthesized within the brain, thus forming a mirror image of the cytokine milieu in the periphery. Although all cells within the brain are capable of initiating cytokine secretion, microglia have an early response to incoming neuronal and humoral stimuli. Inhibition of proinflammatory cytokines that are induced following bacterial infection blocks the appearance of sickness behaviors. Collectively, these data are consistent with the notion that the immune system communicates with the brain to regulate behavior in a way that is consistent with animal survival.


Asunto(s)
Conducta Animal , Encéfalo/fisiopatología , Depresión/etiología , Sistema Inmunológico/fisiopatología , Infecciones/fisiopatología , Neuroinmunomodulación , Animales , Encéfalo/inmunología , Citocinas/inmunología , Depresión/inmunología , Depresión/fisiopatología , Humanos , Sistema Inmunológico/inmunología , Infecciones/complicaciones , Infecciones/inmunología
14.
J Neuroinflammation ; 8: 12, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21306618

RESUMEN

Exogenous administration of insulin-like growth factor (IGF)-I has anti-depressant properties in rodent models of depression. However, nothing is known about the anti-depressant properties of IGF-I during inflammation, nor have mechanisms by which IGF-I alters behavior following activation of the innate immune system been clarified. We hypothesized that central IGF-I would diminish depressive-like behavior on a background of an inflammatory response and that it would do so by inducing expression of the brain-derived neurotrophic factor (BDNF) while decreasing pro-inflammatory cytokine expression in the brain. IGF-I (1,000 ng) was administered intracerebroventricularly (i.c.v.) to CD-1 mice. Mice were subsequently given lipopolysaccharide i.c.v. (LPS, 10 ng). Sickness and depressive-like behaviors were assessed followed by analysis of brain steady state mRNA expression. Central LPS elicited typical transient signs of sickness of mice, including body weight loss, reduced feed intake and decreased social exploration toward a novel juvenile. Similarly, LPS increased time of immobility in the tail suspension test (TST). Pretreatment with IGF-I or antidepressants significantly decreased duration of immobility in the TST in both the absence and presence of LPS. To elucidate the mechanisms underlying the anti-depressant action of IGF-I, we quantified steady-state mRNA expression of inflammatory mediators in whole brain using real-time RT-PCR. LPS increased, whereas IGF-I decreased, expression of inflammatory markers interleukin-1ß (IL-1ß), tumor necrosis factor-(TNF)α, inducible nitric oxide synthase (iNOS) and glial fibrillary acidic protein (GFAP). Moreover, IGF-I increased expression of BDNF. These results indicate that IGF-I down regulates glial activation and induces expression of an endogenous growth factor that shares anti-depressant activity. These actions of IGF-I parallel its ability to diminish depressive-like behavior.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Citocinas/inmunología , Depresión/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Depresión/inmunología , Ingestión de Alimentos/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/inmunología , Lipopolisacáridos/farmacología , Masculino , Ratones , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Pruebas Neuropsicológicas , ARN Mensajero/metabolismo
15.
J Neuroinflammation ; 8: 179, 2011 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-22189158

RESUMEN

Centrally administered insulin-like growth factor (IGF)-I has anti-depressant activity in several rodent models, including lipopolysaccharide (LPS)-induced depression. In this study we tested the ability of IGF-I and GPE (the N-terminal tri-peptide derived from IGF-I) to alter depression-like behavior induced by intraperitoneal (i.p.) administration of LPS in a preventive and curative manner. In the first case, IGF-I (1 µg) or GPE (5 µg) was administered i.c.v. to CD-1 mice followed 30 min later by 330 µg/kg body weight i.p. LPS. In the second case, 830 µg/kg body weight LPS was given 24 h prior to either IGF-I or GPE. When administered i.p., LPS induced full-blown sickness assessed as a loss of body weight, decrease in food intake and sickness behavior. None of these indices were affected by IGF-I or GPE. LPS also induced depression-like behavior; assessed as an increased duration of immobility in the tail suspension and forced swim tests. When administered before or after LPS, IGF-I and GPE abrogated the LPS response; attenuating induction of depression-like behaviors and blocking preexistent depression-like behaviors. Similar to previous work with IGF-I, GPE decreased brain expression of cytokines in response to LPS although unlike IGF-I, GPE did not induce the expression of brain-derived neurotrophic factor (BDNF). LPS induced expression of tryptophan dioxygenases, IDO1, IDO2 and TDO2, but expression of these enzymes was not altered by GPE. Thus, both IGF-I and GPE elicit specific improvement in depression-like behavior independent of sickness, an action that could be due to their anti-inflammatory properties.


Asunto(s)
Conducta Animal/efectos de los fármacos , Depresión/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Lipopolisacáridos/farmacología , Péptidos/farmacología , Péptidos/uso terapéutico , Animales , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Citocinas/genética , Citocinas/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inyecciones Intraperitoneales , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Péptidos/metabolismo
16.
Front Immunol ; 12: 747780, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867973

RESUMEN

Regulatory B cells (Breg) are IL-10 producing subsets of B cells that contribute to immunosuppression in the tumor microenvironment (TME). Breg are elevated in patients with lung cancer; however, the mechanisms underlying Breg development and their function in lung cancer have not been adequately elucidated. Herein, we report a novel role for Indoleamine 2, 3- dioxygenase (IDO), a metabolic enzyme that degrades tryptophan (Trp) and the Trp metabolite L-kynurenine (L-Kyn) in the regulation of Breg differentiation in the lung TME. Using a syngeneic mouse model of lung cancer, we report that Breg frequencies significantly increased during tumor progression in the lung TME and secondary lymphoid organs, while Breg were reduced in tumor-bearing IDO deficient mice (IDO-/-). Trp metabolite L-Kyn promoted Breg differentiation in-vitro in an aryl hydrocarbon receptor (AhR), toll-like receptor-4-myeloid differentiation primary response 88, (TLR4-MyD88) dependent manner. Importantly, using mouse models with conditional deletion of IDO in myeloid-lineage cells, we identified a significant role for immunosuppressive myeloid-derived suppressor cell (MDSC)-associated IDO in modulating in-vivo and ex-vivo differentiation of Breg. Our studies thus identify Trp metabolism as a therapeutic target to modulate regulatory B cell function during lung cancer progression.


Asunto(s)
Linfocitos B Reguladores/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Diferenciación Celular/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Microambiente Tumoral/inmunología , Animales , Carcinoma Pulmonar de Lewis/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ratones , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Triptófano/metabolismo
17.
Brain Behav Immun Health ; 7: 100116, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34589873

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) with pathological features of inflammation, demyelination, and neurodegeneration. Several lines of evidence suggest that the enzymes indoleamine 2,3-dioxygenase (Ido)1 and/or Ido2 influences susceptibility to autoimmune diseases. Deletion of Ido1 exacerbates experimental autoimmune encephalomyelitis (EAE) an animal model of MS. However, no data exist on the role of Ido2 in the pathogenesis of EAE. We investigated whether deletion of Ido2 affected the pathogenesis of EAE. Temporal expression of interferon gamma (Ifng), Ido1 variants, Ido2 variants, as well as genes encoding enzymes of the kynurenine pathway in the spleen and spinal cord of C57BL/6 mice with or without EAE were determined by RT-qPCR. Moreover, EAE was induced in C57BL/6, two Ido1 knockout strains (Ido1KO and Ido1TK) and one Ido2 knockout mouse strain (Ido2-/-) and disease monitored by clinical scores and weight change. Performance on the rotarod was performed on days 0, 5, 10 and 15 post induction. The extent of demyelination in the spinal cord was determined after staining with Oil red O. The development of EAE altered gene expression in both the spleen and spinal cord. Deletion of Ido1 exacerbated the clinical symptoms of EAE. In stark contrast, EAE in Ido2-/- mice did not differ clinically or histologically from control mice. These results confirm a protective role for Ido1, on the pathogenesis of MOG35-55-induced EAE in C57BL/6J mice.

18.
Front Immunol ; 11: 1185, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32612606

RESUMEN

Indoleamine 2, 3-dioxygenase 1 (IDO; IDO1; INDO) is a rate-limiting enzyme that metabolizes the essential amino acid, tryptophan, into downstream kynurenines. Canonically, the metabolic depletion of tryptophan and/or the accumulation of kynurenine is the mechanism that defines how immunosuppressive IDO inhibits immune cell effector functions and/or facilitates T cell death. Non-canonically, IDO also suppresses immunity through non-enzymic effects. Since IDO targeting compounds predominantly aim to inhibit metabolic activity as evidenced across the numerous clinical trials currently evaluating safety/efficacy in patients with cancer, in addition to the recent disappointment of IDO enzyme inhibitor therapy during the phase III ECHO-301 trial, the issue of IDO non-enzyme effects have come to the forefront of mechanistic and therapeutic consideration(s). Here, we review enzyme-dependent and -independent IDO-mediated immunosuppression as it primarily relates to glioblastoma (GBM); the most common and aggressive primary brain tumor in adults. Our group's recent discovery that IDO levels increase in the brain parenchyma during advanced age and regardless of whether GBM is present, highlights an immunosuppressive synergy between aging-increased IDO activity in cells of the central nervous system that reside outside of the brain tumor but collaborate with GBM cell IDO activity inside of the tumor. Because of their potential value for the in vivo study of IDO, we also review current transgenic animal modeling systems while highlighting three new constructs recently created by our group. This work converges on the central premise that maximal immunotherapeutic efficacy in subjects with advanced cancer requires both IDO enzyme- and non-enzyme-neutralization, which is not adequately addressed by available IDO-targeting pharmacologic approaches at this time.


Asunto(s)
Neoplasias Encefálicas/inmunología , Encéfalo/enzimología , Glioblastoma/inmunología , Tolerancia Inmunológica/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Animales , Neoplasias Encefálicas/enzimología , Modelos Animales de Enfermedad , Glioblastoma/enzimología , Humanos , Neoplasias/enzimología , Neoplasias/inmunología
19.
Psychopharmacology (Berl) ; 201(4): 549-56, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18825372

RESUMEN

RATIONALE: During the course of an infection, the pro-inflammatory cytokine tumor necrosis factor alpha (TNFalpha) acts in the brain to trigger development of behavioral responses, collectively termed sickness behavior. Biological activities of TNFalpha can be mediated by TNF receptor type 1 (TNF-R1) and type 2 (TNF-R2). TNFalpha activates neutral sphingomyelinase through the TNF-R1 adapter protein FAN (factor associated with neutral sphingomyelinase activation), but a behavioral role of FAN in the brain has never been reported. OBJECTIVES: We hypothesized that TNFalpha-induced sickness behavior requires TNF-R1 and that FAN is a necessary component for this response. MATERIALS AND METHODS: We determined the role of brain TNF-R1 in sickness behavior by administering an optimal amount of TNFalpha intracerebroventricularly (i.c.v., 50 ng/mouse) to wild-type (WT), TNF-R1-, TNF-R2-, and FAN-deficient mice. Sickness was assessed by decreased social exploration of a novel juvenile, induction of immobility, and loss of body weight. RESULTS: TNF-R1-deficient mice were resistant to the sickness-inducing properties of i.c.v. TNFalpha, whereas both TNF-R2-deficient and WT mice were fully responsive. Furthermore, the complete absence of TNFalpha-induced sickness behavior in FAN-deficient mice provided in vivo evidence that FAN-dependent TNF-R1 signaling is critical for this central action of TNFalpha. CONCLUSIONS: This is the first report to demonstrate that TNFalpha-induced sickness behavior is fully mediated by TNF-R1 and that the adaptor protein FAN is a necessary intracellular intermediate for sickness behavior.


Asunto(s)
Conducta Animal , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Conducta Exploratoria , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/administración & dosificación , Pérdida de Peso
20.
Behav Brain Res ; 359: 731-736, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30243767

RESUMEN

Recent data has supported a role for the gut microbiota in improving cognition and shaping behavior. Here, we assessed whether pectin, a soluble, fermentable fiber, could enhance learning and memory in mice. Two cohorts of young male C57Bl/6 J mice, C1 (n = 20) and C2 (n = 20), were obtained from Jackson Laboratory and randomized to semi-purified AIN-93 M diets containing 5% pectin (n = 10) or cellulose (n = 10). After 16 weeks, learning and memory was assessed by Morris Water Maze (MWM) and microbiota composition was analyzed by 16S rRNA sequencing. Despite identical treatment, we observed differences in learning and memory abilities between cohorts, along with distinct microbiotas. In C1, pectin-fed mice spent a higher percentage of time in the target quadrant at the 24-h probe trial of the MWM versus cellulose-fed mice; in C2, no effect of pectin was observed. In both cohorts, UniFrac distance revealed significant differences in gut microbial communities between cellulose-fed and pectin-fed mice. UniFrac analysis also revealed significantly different bacterial communities between cohorts. Further analysis demonstrated that the microbial genera Oscillospira, Bilophila, and Peptostreptococcoceae were more abundant in C1 versus C2, and positively associated with distance from the platform during the 24-h probe test. These data support previous findings that differences in the gut microbiota may play a role in host response to a dietary intervention and could partly explain irreproducibility in psychological and behavioral experiments. Further research is needed to determine if a causal relationship exists.


Asunto(s)
Fibras de la Dieta/administración & dosificación , Microbioma Gastrointestinal , Aprendizaje por Laberinto/fisiología , Animales , Estudios de Cohortes , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA