Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Intervalo de año de publicación
1.
Cells ; 12(13)2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37443769

RESUMEN

The term 'perinatal environment' refers to the period surrounding birth, which plays a crucial role in brain development. It has been suggested that dynamic communication between the neuro-immune system and gut microbiota is essential in maintaining adequate brain function. This interaction depends on the mother's status during pregnancy and/or the newborn environment. Here, we show experimental and clinical evidence that indicates that the perinatal period is a critical window in which stress-induced immune activation and altered microbiota compositions produce lasting behavioral consequences, although a clear causative relationship has not yet been established. In addition, we discuss potential early treatments for preventing the deleterious effect of perinatal stress exposure. In this sense, early environmental enrichment exposure (including exercise) and melatonin use in the perinatal period could be valuable in improving the negative consequences of early adversities. The evidence presented in this review encourages the realization of studies investigating the beneficial role of melatonin administration and environmental enrichment exposure in mitigating cognitive alteration in offspring under perinatal stress exposure. On the other hand, direct evidence of microbiota restoration as the main mechanism behind the beneficial effects of this treatment has not been fully demonstrated and should be explored in future studies.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo , Disfunción Cognitiva , Exposición Materna , Efectos Tardíos de la Exposición Prenatal , Estrés Psicológico , Disfunción Cognitiva/inmunología , Disfunción Cognitiva/microbiología , Disfunción Cognitiva/prevención & control , Humanos , Femenino , Animales , Efectos Tardíos de la Exposición Prenatal/etiología , Melatonina/administración & dosificación , Encéfalo/crecimiento & desarrollo , Neurogénesis , Antioxidantes/administración & dosificación , Probióticos/administración & dosificación
2.
Pharmacol Res Perspect ; 9(5): e00795, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34609083

RESUMEN

Neurodegenerative diseases (NDD) are disorders characterized by the progressive loss of neurons affecting motor, sensory, and/or cognitive functions. The incidence of these diseases is increasing and has a great impact due to their high morbidity and mortality. Unfortunately, current therapeutic strategies only temporarily improve the patients' quality of life but are insufficient for completely alleviating the symptoms. An interaction between the immune system and the central nervous system (CNS) is widely associated with neuronal damage in NDD. Usually, immune cell infiltration has been identified with inflammation and is considered harmful to the injured CNS. However, the immune system has a crucial role in the protection and regeneration of the injured CNS. Nowadays, there is a consensus that deregulation of immune homeostasis may represent one of the key initial steps in NDD. Dr. Michal Schwartz originally conceived the concept of "protective autoimmunity" (PA) as a well-controlled peripheral inflammatory reaction after injury, essential for neuroprotection and regeneration. Several studies suggested that immunizing with a weaker version of the neural self-antigen would generate PA without degenerative autoimmunity. The development of CNS-related peptides with immunomodulatory neuroprotective effect led to important research to evaluate their use in chronic and acute NDD. In this review, we refer to the role of PA and the potential applications of active immunization as a therapeutic option for NDD treatment. In particular, we focus on the experimental and clinical promissory findings for CNS-related peptides with beneficial immunomodulatory effects.


Asunto(s)
Autoantígenos/uso terapéutico , Autoinmunidad/inmunología , Factores Inmunológicos/uso terapéutico , Regeneración Nerviosa/inmunología , Enfermedades Neurodegenerativas/terapia , Neuroprotección/inmunología , Péptidos/uso terapéutico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/terapia , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/terapia , Animales , Acetato de Glatiramer/uso terapéutico , Humanos , Inmunización Pasiva , Inmunomodulación , Proteína Básica de Mielina/uso terapéutico , Enfermedades Neurodegenerativas/inmunología , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/terapia , Fragmentos de Péptidos/uso terapéutico , Deficiencias en la Proteostasis , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/terapia , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/terapia
3.
Stress ; 24(6): 987-997, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34581257

RESUMEN

During gestation, stress exposure increases the risk of developing cognitive and physiological alterations in either the long or short term. Among them, metabolic alterations have been described. Adipose tissue is responsible for the secretion of several factors involved in controlling body weight and energy expenditure, the regulation of insulin sensitivity, and the development of inflammation, among others. Moreover, the liver regulates glucose homeostasis and lipid metabolism, playing an essential role in developing insulin resistance. In this work, we analyzed if prenatal stress leads to alterations in metabolism and the relationship between these alterations and gene expression in the adipose tissue and the liver. Prenatal stress-exposed animals developed disturbances in the glucose and insulin response curve, showing in both tests higher glycemia than the control group. However, they did not exhibit increased body weight. At the same time, in the adipose tissue, we observed an increase in mRNA expression of Leptin and Resistin and a decrease in Adiponectin. In the liver, we observed a lower mRNA expression of several genes involved in glucose metabolism and fatty acid oxidation, such as Sirt1, Pgc1α, Pparα, among others. In both tissues, we observed a lower expression of inflammatory genes. These results suggest that prenatal stress exposure produces insulin resistance at both physiological and molecular levels without pro-inflammatory signaling or obesity.


Asunto(s)
Resistencia a la Insulina , Tejido Adiposo/metabolismo , Animales , Femenino , Inflamación/metabolismo , Insulina , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Embarazo , Estrés Psicológico
4.
J Dev Orig Health Dis ; 12(5): 721-730, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33118903

RESUMEN

Prenatal insults during fetal development result in increased likelihood of developing chronic disease. Obesity, the biggest risk factor for the development of metabolic disease, is affected by several genetic and environmental factors. High-fat diet (HFD) consumption is usually linked with the development of obesity. The main goal of this study was to analyze the impact of the exposure to a HFD in prenatally stressed animals. For this purpose, we subjected pregnant BALB/c mice to restraint stress for 2 h a day between gestational day (GD) 14 and GD 21. Prenatally stressed and control offspring of both sexes were postnatally exposed to a HFD for 24 weeks. We found that prenatal stress (PS) per se produced disturbances in males such as increased total blood cholesterol and triglycerides, with a decrease in mRNA expression of sirtuin-1. When these animals were fed a HFD, we observed a rise in glucose and insulin levels and an increase in visceral adipose tissue gene expression of leptin, resistin, and interleukin-1 beta. Although females proved to be more resilient to PS consequences, when they were fed a HFD, they showed significant metabolic impairment. In addition to the changes observed in males, females also presented an increase in body weight and adiposity and a rise in cholesterol levels.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Enfermedades Metabólicas/etiología , Ratones Endogámicos BALB C/metabolismo , Animales , Dieta Alta en Grasa/métodos , Modelos Animales de Enfermedad , Femenino , Enfermedades Metabólicas/dietoterapia , Ratones , Ratones Endogámicos BALB C/anomalías , Embarazo
5.
Clin Ther ; 42(6): 993-1006, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32224031

RESUMEN

PURPOSE: Chronic and persistent exposure to negative stress can lead to adverse consequences on health. Particularly, psychosocial factors were found to increase the risk and outcome of respiratory diseases like asthma. Glucocorticoids (GCs) are the most efficient anti-inflammatory therapy for asthma. However, a significant proportion of patients don't respond adequately to GC administration. GC sensitivity is modulated by genetic and acquired disease-related factors. Additionally, it was proposed that endogenous corticosteroids may limit certain actions of synthetic GCs, contributing to insensitivity. Psychological and physiological stresses activate the hypothalamic-pituitary-adrenal axis, increasing cortisol levels. Here, we review the mechanism involved in altered GC sensitivity in asthmatic patients under stressful situations. Strategies for modulation GC sensitivity and improving GC therapy are discussed. METHODS: PubMed was searched for publications on psychological chronic stress and asthma, GC resistance in asthma, biological mechanisms for GC resistance, and drugs for steroid-resistant asthma, including highly potent GCs. FINDINGS: GC resistance in patients with severe disease remains a major clinical problem. In asthma, experimental and clinical evidence suggests that chronic stress induces inflammatory changes, contributing to a worse GC response. GC resistant patients can be treated with other broad-spectrum anti-inflammatory drugs, but these generally have major side effects. Different mechanisms of GC resistance have been described and might be useful for developing new therapeutic strategies against it. Novel drugs, such as highly potent GCs, phosphoinositide 3-kinase-delta inhibitors that reestablish histone deacetylase-2 function, decrease of GC receptor phosphorylation by p38 mitogen-activated protein kinase inhibitors, or phosphatase activators, are currently in clinical development and might be combined with GC therapy in the future. Furthermore, microRNAs (small noncoding RNA molecules) operate as posttranscriptional regulators, providing another level of control of GC receptor levels. Empirical results allow postulating that the detection and study of microRNAs might be a promising approach to better characterize and treat asthmatic patients. IMPLICATIONS: Many molecular and cellular pathobiological mechanisms are responsible of GC resistance. Therefore detecting specific biomarkers to help identify patients who would benefit from new therapies is crucial. Stress consitutes a negative aspect of current lifestyles that increase asthma morbidity and mortality. Adequate stress management could be an important and positive intervention.


Asunto(s)
Asma/tratamiento farmacológico , Resistencia a Medicamentos , Glucocorticoides/uso terapéutico , Receptores de Glucocorticoides/inmunología , Estrés Psicológico/complicaciones , Animales , Asma/inmunología , Enfermedad Crónica , Humanos , Estrés Psicológico/inmunología
6.
Front Immunol ; 9: 1341, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29971064

RESUMEN

Clinical data and experimental studies have suggested a relationship between psychosocial factors and cancer prognosis. Both, stress effects on the immune system and on tumor biology were analyzed independently. However, there are few studies regarding the stress influence on the interplay between the immune system and tumor biology. Moreover, antidepressants have been used in patients with cancer to alleviate mood disorders. Nevertheless, there is contradictory evidence about their action on cancer prognosis. In this context, we investigated the effect of chronic stress on tumor progression taking into account both its influence on the immune system and on tumor biology. Furthermore, we analyzed the action of selective serotonin reuptake inhibitors, fluoxetine and sertraline, in these effects. For this purpose, C57BL/6J mice submitted or not to a chronic stress model and treated or not with fluoxetine or sertraline were subcutaneously inoculated with EL4 cells to develop solid tumors. Our results indicated that chronic stress leads to an increase in both tumor growth and tumor cell dissemination. The analysis of cell cycle regulatory proteins showed that stress induced an increase in the mRNA levels of cyclins A2, D1, and D3 and a decrease in mRNA levels of cell cycle inhibitors p15, p16, p21, p27, stimulating cell cycle progression. Moreover, an augment of mRNA levels of metalloproteases (MMP-2 and MMP-9), a decrease of inhibitors of metalloproteases mRNA levels (TIMP 1, 2, and 3), and an increase in migration ability were found in tumors from stressed animals. In addition, a significant decrease of antitumor immune response in animals under stress was found. Adoptive lymphoid cell transfer experiments indicated that the reduced immune response in stressed animals influenced both the tumor growth and the metastatic capacity of tumor cells. Finally, we found an important beneficious effect of fluoxetine or sertraline treatment on cancer progression. Our results emphasize the crucial role of the immune system in tumor progression under stress situations. Although a direct effect of stress and drug treatment on tumor biology could not be ruled out, the beneficial effect of fluoxetine and sertraline appears to be mainly due to a restoration of antitumor immune response.

7.
Cytokine ; 107: 26-34, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29175260

RESUMEN

It is known that long-term exposure to stressful situations can produce severe consequences affecting behavioral, endocrine and immunological parameters. We have previously shown that stressed BALB/c mice had poor learning performance, which was reverted by glatiramer acetate treatment through a mechanism that likely involved the regulation of the cytokine balance and adult neurogenesis. In addition, recent results suggest that cytokine and neurotrophin expression in the hippocampus displayed similar tendencies as those in the serum. However, if lymphoid cells could be good candidates as peripheral markers of memory impairment have not yet been investigated. For this purpose, we analyzed the spatial memory and the neutrophin and cytokine mRNA levels in lymph nodes and hippocampus in mice submitted to chronic stress treated or not with glatiramer acetate. Results indicated that there was a correlation between the cytokine and neurotrophin mRNA levels in the hippocampus and in the peripheral lymph nodes, and the cognitive performance in BALB/c mice. In particular, our results suggest that altered IFN-γ levels could be used as peripheral biomarker of cognitive deficit and treatment response.


Asunto(s)
Biomarcadores/metabolismo , Trastornos del Conocimiento/metabolismo , Interferón gamma/metabolismo , Linfocitos/metabolismo , Animales , Biomarcadores/sangre , Enfermedad Crónica , Trastornos del Conocimiento/sangre , Trastornos del Conocimiento/fisiopatología , Femenino , Expresión Génica/efectos de los fármacos , Acetato de Glatiramer/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Interferón gamma/sangre , Interferón gamma/genética , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/metabolismo , Linfocitos/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Endogámicos BALB C , Factores de Crecimiento Nervioso/sangre , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Estrés Psicológico/fisiopatología , Estrés Psicológico/prevención & control
9.
Pharmacol Res ; 109: 101-7, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26644208

RESUMEN

Selective serotonin reuptake inhibitors are frequently used antidepressants. In particular, fluoxetine is usually chosen for the treatment of the symptoms of depression, obsessive-compulsive, panic attack and bulimia nervosa. Antidepressant therapy has been associated with immune dysfunction. However, there is contradictory evidence about the effect of fluoxetine on the immune system. Experimental findings indicate that lymphocytes express the serotonin transporter. Moreover it has been shown that fluoxetine is able to modulate the immune function through a serotonin-dependent pathway and through a novel independent mechanism. In addition, several studies have shown that fluoxetine can alter tumor cell viability. Thus, it was recently demonstrated in vivo that chronic fluoxetine treatment inhibits tumor growth by increasing antitumor T-cell activity. Here we briefly review some of the literature referring to how fluoxetine is able to modify, for better or worse, the functionality of the immune system. These results of our analysis point to the relevance of the novel pharmacological action of this drug as an immunomodulator helping to treat several pathologies in which immune deficiency and/or deregulation is present.


Asunto(s)
Antidepresivos/farmacología , Fluoxetina/farmacología , Factores Inmunológicos/farmacología , Animales , Antidepresivos/uso terapéutico , Fluoxetina/uso terapéutico , Homeostasis/efectos de los fármacos , Humanos , Factores Inmunológicos/uso terapéutico , Serotonina/metabolismo
10.
Psychopharmacology (Berl) ; 232(9): 1595-605, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25373868

RESUMEN

RATIONALE: Oxidative stress and neurotrophins are among the most important factors involved in several pathophysiological brain processes. In addition, long-term exposure to stressful situations has deleterious effects on behaviour. We have previously shown that stressed female BALB/c mice show poor learning performance and that this behaviour is reversed by glatiramer acetate (GA) treatment. OBJECTIVES: We investigated the involvement of the hippocampal oxidative status and neurotrophin levels in cognitive deficit and the improvement of this deficit by GA treatment in chronic stressed BALB/c mice. METHODS: Female BALB/c mice were exposed to a chronic mild stress (CMS) model for 9 weeks. During the last 3 weeks of the stress exposure, one group of mice was subcutaneously injected four times with 100 µg GA/mouse. Following this period, behavioural studies were performed. The mice were then sacrificed, and biochemical studies were performed on the hippocampus. RESULTS: The stressed mice exhibited a significant decline in their performance in the open-field and in object-in-place tasks. This decline was accompanied by an increase in reactive oxygen species (ROS) and a decrease in nitric oxide (NO) production by neuronal nitric oxide synthase (nNOS). Neither antioxidant defences nor neurotrophin protein levels were involved in this process. Interestingly, the administration of GA re-established the normal levels of ROS, restored nNOS activity and improved learning performance. CONCLUSIONS: The GA treatment improved learning and memory in female BALB/c mice under chronic stress through a mechanism that involves the regulation of NO production, which in turn modulates the ROS levels.


Asunto(s)
Acetato de Glatiramer/farmacología , Hipocampo/metabolismo , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Óxido Nítrico/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Femenino , Hipocampo/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Factores de Crecimiento Nervioso/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Estrés Oxidativo/fisiología , Estrés Psicológico/complicaciones
11.
Physiol Behav ; 128: 141-7, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24518864

RESUMEN

It has been demonstrated that a short-duration stress (acute stress) may result in immunopreparatory or immunoenhancing physiological conditions. The aim of the present study was to investigate whether exposure to prenatal restraint stress (PRS) influences the impact of acute stress on the T-cell response in the adult life. We found that female mice exposed to PRS (PS mice) did not exhibit changes in the T-cell-dependent IgG antibody production with respect to prenatally non-stressed mice (no-PS mice). However, no-PS mice exposed to acute stress showed an increase of antibody production after antigen stimulation. In contrast, PS mice exhibited a decreased response after an acute situation. Spleen catecholamines and plasma corticosterone levels were increased in acute stress in both PS and no-PS mice. Nevertheless, lymphocyte response to hormones was altered in PS mice. Particularly, inhibitory effect of corticosterone was higher on lymphocytes from PS mice. In addition, an increase in protein levels and mRNA expression of glucocorticoid receptor was found in lymphoid cells from PS mice. These results show that prenatal stress alters the immune intrinsic regulatory mechanism that in turn induces an increased vulnerability to any stressful situation able to modify immune homeostasis.


Asunto(s)
Linfocitos/fisiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Receptores de Glucocorticoides/fisiología , Estrés Psicológico/fisiopatología , Linfocitos T/fisiología , Animales , Corticosterona/sangre , Epinefrina/análisis , Epinefrina/fisiología , Femenino , Inmunidad Celular/inmunología , Inmunidad Celular/fisiología , Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Norepinefrina/análisis , Norepinefrina/fisiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inmunología , Restricción Física/efectos adversos , Bazo/química , Bazo/fisiopatología , Estrés Psicológico/inmunología , Regulación hacia Arriba/inmunología , Regulación hacia Arriba/fisiología
12.
Open Cardiovasc Med J ; 7: 90-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24155799

RESUMEN

The pathogenesis of atherosclerosis includes the assignment of a critical role to cells of the monocyte/macrophage lineage and to pro-inflammatory cytokines. Niacin is known to improve lipid metabolism and to produce beneficial modification of cardiovascular risk factors. The aim of this work was to investigate if Niacin is able to modulate pro-inflammatory cytokine production in macrophages in a murine model of atherosclerosis. For this purpose C57Bl/6J mice fed with atherogenic diet (AGD) or with conventional chow diet were used. The AGD group showed an increase in body weight and in total plasma cholesterol, with no differences in triglyceride or HDL levels. Lesions in arterial walls were observed. The characterization of Niacin receptor showed an increase in the receptor number of macrophages from the AGD group. Macrophages from control and AGD animals treated in vitro with an inflammatory stimulus showed elevated levels of IL-6, IL-1 and TNF-α, that were even higher in macrophages from AGD mice. Niacin was able to decrease the production of pro-inflammatory cytokines in stimulated macrophages. Similar effect of Niacin was observed in an in vivo model of inflammation. These results show an attenuating inflammatory mechanism for this therapeutic agent and would point out its potential action in plaque stabilization and in the prevention of atherosclerosis progression. Furthermore, the present results provide the basis for future studies on the potential contribution of Niacin to anti-inflammatory therapies.

13.
Brain Behav Immun ; 26(3): 429-38, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22200600

RESUMEN

Long-term exposure to stressful situations has deleterious effects on adult neurogenesis, behavior, and the immune system. We have previously shown that stressed BALB/c mice show poor learning performance, which correlates with an increase in the T helper 1/T helper 2 (Th1/Th2) cytokine balance. Glatiramer acetate (GA) can stimulate autoreactive T cells. In this work we investigated the effects of GA treatment on BALB/c mice exposed to chronic mild stress (CMS). Stressed mice exhibited a significant decline in their performance in the open field and Y-maze tasks, which was accompanied by a reduction in dentate gyrus neurogenesis and an altered Th1/Th2 balance. Interestingly, after 6 weeks of CMS exposure administration of GA reestablished normal levels of adult neurogenesis, restored the Th1/Th2 balance, and improved learning performance. These results demonstrate that GA treatment can reverse the learning impairment induced by stress through a mechanism that likely involves the regulation of the cytokine balance and adult neurogenesis.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Hipocampo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Péptidos/farmacología , Estrés Psicológico/inmunología , Balance Th1 - Th2/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Citocinas/efectos de los fármacos , Femenino , Acetato de Glatiramer , Ratones , Ratones Endogámicos BALB C
14.
Eur J Pharmacol ; 659(2-3): 265-72, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21497159

RESUMEN

Antidepressants have a controversial role with regard to their influence on cancer and immunity. Recently, we showed that fluoxetine administration induces an enhancement of the T-cell mediated immunity in naïve mice, resulting in the inhibition of tumor growth. Here we studied the effects of fluoxetine on lymphoma proliferation/apoptosis and immunity in tumor bearing-mice. We found an increase of apoptotic cells (active Caspase-3(+)) and a decrease of proliferative cells (PCNA(+)) in tumors growing in fluoxetine-treated animals. In addition, differential gene expressions of cell cycle and death markers were observed. Cyclins D3, E and B were reduced in tumors from animals treated with fluoxetine, whereas the tumor suppressor p53 and the cell cycle inhibitors p15/INK4B, p16/INK4A and p27/Kip1 were increased. Besides, the expression of the antiapoptotic factor Bcl-2 and the proapoptotic factor Bad were lower and higher respectively in these animals. These changes were accompanied by increased IFN-γ and TNF-α levels as well as augmented circulating CD8(+) T lymphocytes in tumor-bearing mice treated with the antidepressant. Therefore, we propose that the up-regulation of T-cell mediated antitumor immunity may be contributing to the alterations of tumor cell proliferation and apoptosis thus resulting in the inhibition of tumor progression.


Asunto(s)
Apoptosis/efectos de los fármacos , Fluoxetina/administración & dosificación , Fluoxetina/farmacología , Linfoma/inmunología , Linfoma/patología , Linfocitos T/inmunología , Regulación hacia Arriba/efectos de los fármacos , Administración Oral , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Linfoma/diagnóstico , Linfoma/genética , Ratones , Ratones Endogámicos BALB C , Pronóstico , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Linfocitos T/efectos de los fármacos , Regulación hacia Arriba/inmunología
15.
Cancer Invest ; 29(1): 29-36, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21166496

RESUMEN

Larrea divaricata exerts a dual effect on the proliferation of BW5147 cells. The aim of this work was to analyze the effect of leukotrienes on the antiproliferative action of the extract in relation to cyclic adenosine monophosphate production and protein kinase C activation. The extracts induced cyclic adenosine monophosphate and inhibited protein kinase C but induced leukotriene B4 production, which exerted contrary effects. These results suggest the finding of an extract free of compounds, which can increase leukotriene B4 level, in order to obtain a more active extract to be used in the treatment of lymphomas, positively modulated by leukotrienes.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proliferación Celular/efectos de los fármacos , GMP Cíclico/metabolismo , Larrea , Leucotrieno B4/metabolismo , Linfoma de Células T/enzimología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Activación Enzimática , Linfoma de Células T/patología , Ratones , Extractos Vegetales/farmacología , Hojas de la Planta , Proteína Quinasa C/metabolismo , Regulación hacia Arriba
16.
Biol Psychiatry ; 65(11): 935-42, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19167703

RESUMEN

BACKGROUND: Stress alters the neuroendocrine system, immunity, and cancer. Although the classic stress hormones are glucocorticoids and catecholamines, thyroid hormones have also been related to stress. We recently reported that chronic restraint stress impairs T-cell mediated immunity and enhances tumor growth in mice. METHODS: To study the participation of these hormones on the stress-induced alterations of the immune function and lymphoma growth, mice were subjected to acute or chronic stress, with or without thyroxin supplementation. Hormone levels, immune status, and cancer progression were evaluated. RESULTS: Differential endocrine alterations were observed in response to acute and chronic stress. Although corticosterone and noradrenaline levels were increased by acute stress, they were restored after prolonged exposure to the stressor. Instead, thyroid hormone levels were only reduced in chronically stressed animals in comparison with control subjects. Correlating, chronic but not acute stress impaired T-cell reactivity. Thyroxin replacement treatment of chronic restraint stress-exposed mice, which restored the euthyroid status, reversed the observed reduction of T-cell lymphoproliferative responses. Moreover, therapeutic thyroid replacement also reversed the alterations of lymphoma growth induced by chronic stress in syngeneic mice bearing tumors as well as Interleukin-2 production and specific cytotoxic response against tumor cells. Finally, we found that the isoforms theta and alpha of the protein kinase C are involved in these events. CONCLUSIONS: These results show for the first time that thyroid hormones are important neuroendocrine regulators of tumor evolution, most probably acting through the modulation of T-cell mediated immunity affected by chronic stress.


Asunto(s)
Linfoma/etiología , Estrés Psicológico/inmunología , Estrés Psicológico/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Concanavalina A/farmacología , Corticosterona/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Linfoma/inmunología , Linfoma/patología , Ratones , Ratones Endogámicos BALB C , Mitógenos/farmacología , Norepinefrina/metabolismo , Proteína Quinasa C/metabolismo , Restricción Física/métodos , Estrés Psicológico/complicaciones , Estrés Psicológico/tratamiento farmacológico , Timidina/metabolismo , Hormonas Tiroideas/administración & dosificación , Tiroxina/farmacología , Tritio/metabolismo
17.
Stress ; 12(4): 350-61, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19006005

RESUMEN

Nitric oxide (NO) has been involved in many pathophysiological brain processes. Recently, we showed that neuronal nitric oxide synthase (nNOS)-mediated decrease in NO production is involved in memory impairment induced by chronic mild stress (CMS) in BALB/c mice. Two genetically different inbred murine strains, C57BL/6 and BALB/c, show distinct behavioral responses, neurodevelopmental and neurochemical parameters. Here, we perform a comparative study on CMS effects upon learning and memory in both strains, analyzing the role of NO production and its regulation by protein kinase C (PKC). Stressed BALB/c, but not C57Bl/6 mice, showed a poor learning performance in both the open field and passive avoidance inhibitory tasks. Also, CMS induced a diminished NO production by nNOS, associated with an increment in gamma and zeta PKC isoenzymes in BALB/c mice. In C57BL/6 mice, CMS had no effect on NO production, but increased delta and decreased betaI PKC isoforms. In vivo administration of a NOS inhibitor induced behavioral alterations in both strains. These results suggest a differential effect of stress, with BALB/c being more vulnerable to stress than C57BL/6 mice. This effect could be related to a differential regulation of NOS and PKC isoenzymes, pointing to an important role of NO in learning and memory.


Asunto(s)
Conducta Animal/efectos de los fármacos , Aprendizaje , Memoria , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/biosíntesis , Proteína Quinasa C/metabolismo , Estrés Psicológico/psicología , Animales , Reacción de Prevención , Conducta Exploratoria/efectos de los fármacos , Femenino , Hipocampo/enzimología , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Estrés Psicológico/fisiopatología
18.
Brain Behav Immun ; 23(1): 36-40, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18625298

RESUMEN

Chronic stress and depression are widely known to down-regulate the immune system, and several antidepressants can reverse this impairment, with or without effects in normal subjects. Although the central nervous system is undoubtedly involved in these events, some psychotropic drugs can also exert direct effects on lymphoid cells. We have recently shown that the antidepressant fluoxetine enhances T cell proliferation and T(H)1 cytokine production in vivo, without changes on CD4/CD8 subsets. In vitro, a direct action of fluoxetine upon T lymphocyte reactivity by complex mechanisms was also described. In another work, we also found that chronic stress reduces T cell mediated immunity, namely a decrease of T cell response to mitogens, T(H)1 cytokine production and CD4+-but not CD8+--T lymphocytes. Here we investigated the effects of fluoxetine on chronic stress-driven immune system depression. We found that fluoxetine restored T cell proliferation and interleukin-2, interferon-gamma and tumor necrosis factor-alpha production by compensatory mechanisms. In addition, CD4/CD8 ratio was also normalized by antidepressant administration, but this seems to be a non-compensatory effect associated specifically to stress. No changes were observed in other lymphoid cells, i.e. natural killer cells and B lymphocytes. Finally, we observed that fluoxetine is able to reverse T cell reactivity impairment in vitro by a direct action at clinically relevant doses. These results highlight the relevance of pharmacological treatment of stress and depression, and may help to begin elucidating the complex events triggered--directly and/or indirectly--by antidepressants in non-neuronal cell types.


Asunto(s)
Fluoxetina/uso terapéutico , Estrés Psicológico/prevención & control , Linfocitos T/efectos de los fármacos , Animales , Antidepresivos de Segunda Generación/administración & dosificación , Antidepresivos de Segunda Generación/uso terapéutico , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Relación CD4-CD8 , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Enfermedad Crónica , Femenino , Citometría de Flujo , Fluoxetina/administración & dosificación , Interferón gamma/genética , Interleucina-2/genética , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos BALB C , Restricción Física/efectos adversos , Restricción Física/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Psicológico/etiología , Estrés Psicológico/fisiopatología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/genética
19.
Neuroimmunomodulation ; 15(1): 68-75, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18667802

RESUMEN

Autoimmune thyroid diseases (AITD) are the most common organ-specific autoimmune disorders affecting approximately 5% of the overall population. An aberrant interaction between abnormal thyrocytes, abnormal antigen-presenting cells and abnormal T cells forms the basis for the atypical autoimmune reaction targeting thyroid antigens. It was proposed that nongenetic (environmental and hormonal) factors play a crucial etiological role in AITD development, through altering immune-endocrine interactions. The most outstanding fact is that in genetically predisposed individuals, the disruption of these neuroendocrine-immune interactions by environmental factors results in thyroid autoimmune dysfunction. These interactions are able to incline the balance between Th1-Th2 immune response toward one side, resulting in a Th1-cell-mediated autoimmune reaction with thyrocyte destruction and hypothyroidism in Hashimoto's thyroiditis but to a hyperreactive Th2-mediated humoral response against TSH receptor with stimulatory antibodies leading to Graves' disease hyperthyroidism. In this review the main mechanisms involved are summarized. In this sense, the participation of stress-mediated activation of the sympathoadrenal system and hypothalamic-pituitary-adrenal axis, the hormonal changes occurring during pregnancy and postpartum acting on antigen-presenting cells and influencing, in this way, the balance of the immune status are shown to participate in AITD etiology. The possibility that altered levels of thyroid hormones during the course of the AITD may alter immune function is also discussed.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades del Sistema Endocrino/inmunología , Sistema Inmunológico/inmunología , Enfermedades de la Tiroides/inmunología , Animales , Enfermedades Autoinmunes/fisiopatología , Enfermedades del Sistema Endocrino/fisiopatología , Humanos , Sistema Inmunológico/fisiopatología , Inmunidad Celular/inmunología , Receptores de Tirotropina/inmunología , Estrés Psicológico/inmunología , Estrés Psicológico/fisiopatología , Linfocitos T/inmunología , Enfermedades de la Tiroides/fisiopatología , Glándula Tiroides/inmunología , Glándula Tiroides/fisiopatología
20.
Biochem Pharmacol ; 75(9): 1817-26, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18342838

RESUMEN

Fluoxetine, a selective serotonin reuptake inhibitor, is widely used for the treatment of depressive symptoms of cancer patients. However, there are contradictory evidences about its effects on immunity and cancer. Thus, we studied the effects of fluoxetine on tumor growth and on antitumoral T-cell-mediated immunity. In vivo chronic fluoxetine treatment inhibited tumor growth, and increased latency of appearance of solid tumors and survival of mice. Fluoxetine administration also increased mitogen-induced T-cell proliferation and Tumor Necrosis Factor-alpha (TNF-alpha) and Interferon-gamma (IFN-gamma) expression, without altering CD4(+)/CD8(+) ratio. In vitro, fluoxetine did not affect tumor cells proliferation, but it exerted a direct effect on T lymphocytes. Both fluoxetine and serotonin stimulated proliferation induced by a suboptimal mitogen concentration but inhibited proliferation at the optimal one. When both drugs were combined the results indicated that the effects of fluoxetine are in part independent of its ability to elevate serotonin extracellular levels. Finally, continue fluoxetine administration in nude mice - devoid of T lymphocytes - did not modify tumor progression, thus supporting the hypothesis of an immuno-modulatory effect of this drug on T cells that drives tumor growth control. These findings indicate, for the first time, that fluoxetine inhibits tumor growth through modulation of T-cell-mediated immunity by the already known serotonin-dependent pathway and by a novel independent mechanism.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Fluoxetina/uso terapéutico , Linfoma de Células T/tratamiento farmacológico , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Serotonina/metabolismo , Linfocitos T/efectos de los fármacos , Animales , Relación CD4-CD8 , Línea Celular Tumoral , Citocinas/biosíntesis , Femenino , Fluoxetina/administración & dosificación , Fluoxetina/farmacología , Inmunidad Celular/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Linfoma de Células T/inmunología , Linfoma de Células T/metabolismo , Linfoma de Células T/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Linfocitos T/inmunología , Linfocitos T/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...