Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Menopause ; 25(12): 1424-1431, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29994967

RESUMEN

OBJECTIVE: Cognitive outcomes in trials of postmenopausal hormone treatment have been inconsistent. Differing outcomes may be attributed to hormone formulation, treatment duration and timing, and differential cognitive domain effects. We previously demonstrated treatment benefits on visual cognitive function. In the present study, we describe the effects of hormone treatment on verbal outcomes in the same women, seeking to understand the effects of prior versus current hormone treatment on verbal function. METHODS: This is a cross-sectional evaluation of 57 women (38 hormone users [25 prior long-term users and 13 current users] and 19 never-users). Hormone users took identical formulations of estrogen or estrogen + progestin (0.625 mg/d conjugated equine estrogens with or without medroxyprogesterone acetate) for at least 10 years, beginning within 2 years of menopause. Women were evaluated with tests of verbal function and functional magnetic resonance imaging (fMRI) of a verbal discrimination task. RESULTS: All women scored similarly on assessments of verbal function (Hopkins Verbal Learning Test and a verbal discrimination task performed during the fMRI scanning session); however, women ever treated with hormones had more left inferior frontal (T = 3.72; P < 0.001) and right prefrontal cortex (T = 3.53; P < 0.001) activation during the verbal task. Hormone-treated women performed slightly worse on the verbal discrimination task (mean accuracy 81.72 ±â€Š11.57 ever-treated, 85.30 ±â€Š5.87 never-treated, P = 0.14), took longer to respond (mean reaction time 1.10 ±â€Š0.17 s ever-treated, 1.02 ±â€Š0.11 never-treated, P = 0.03), and remembered fewer previously viewed words (mean accuracy 62.21 ±â€Š8.73 ever-treated, 65.45 ±â€Š7.49 never-treated, P = 0.18). Increased posterior cingulate activity was associated with longer response times (R = 0.323, P = 0.015) and worse delayed verbal recall (R = -0.328, P = 0.048), suggesting that increased activation was associated with less efficient cognitive processing. We did not detect between group differences in activation in the left prefrontal cortex, superior frontal cortex, thalamus, or occipital/parietal junction. CONCLUSIONS: Although current and past hormone treatment was associated with differences in neural pathways used during verbal discrimination, verbal function was not higher than never-users.


Asunto(s)
Cognición/efectos de los fármacos , Moduladores de los Receptores de Estrógeno/farmacología , Terapia de Reemplazo de Estrógeno/psicología , Estrógenos Conjugados (USP)/farmacología , Estrógenos/farmacología , Acetato de Medroxiprogesterona/farmacología , Vías Nerviosas/efectos de los fármacos , Posmenopausia/efectos de los fármacos , Anciano , Estudios Transversales , Combinación de Medicamentos , Femenino , Humanos , Imagen por Resonancia Magnética , Memoria a Corto Plazo/fisiología , Recuerdo Mental/fisiología , Persona de Mediana Edad , Pruebas Neuropsicológicas , Corteza Prefrontal/diagnóstico por imagen , Corteza Prefrontal/metabolismo , Tiempo de Reacción , Resultado del Tratamiento , Aprendizaje Verbal
2.
Psychoneuroendocrinology ; 76: 218-225, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27622993

RESUMEN

Disturbances of emotion regulation and depressive symptoms are common during the menopause transition. Reproductive hormone levels are not directly correlated with depressive symptoms, and other factors may influence mood symptoms during menopause. In this study, we sought to determine the role of metabolic function in mood symptoms during menopause, hypothesizing an association with menopause status and long-term glucose load. We studied 54 women across three menopause transition stages (15 premenopause, 11 perimenopause, and 28 postmenopause), examining effects of age, hormones, and metabolism on mood and neural activation during emotional discrimination. We assessed participants using behavioral and functional MRI measures of negative emotion and emotion discrimination, and glycated hemoglobin A1c, to assess long-term glucose load. We found that emotionally unpleasant images activated emotion regulation (amygdala) and cognitive association brain regions (prefrontal cortex, posterior cingulate, temporal-parietal-occipital (TPO) junction, hippocampus). Cognitive association region activity increased with menopause stage. Perimenopausal women had left TPO junction activation, and postmenopausal women had prefrontal cortex, posterior cingulate, and TPO junction activation. Negative affect was associated with decreased amygdala activation, while depression symptoms and negative mood were associated with increased TPO junction activation. Hemoglobin A1c was associated with negative interpretation bias of neutral images and cognitive region recruitment during emotion discrimination. FSH levels, indicating menopause stage, were associated with negative mood. Age was not associated with any behavioral measures or activation patterns during the emotion task. Our results suggest that an interaction between metabolic and hormonal factors may influence emotion regulation, leading to increased risk for depression during menopause.


Asunto(s)
Síntomas Afectivos , Amígdala del Cerebelo/fisiología , Corteza Cerebral/fisiología , Toma de Decisiones/fisiología , Depresión , Emociones/fisiología , Hemoglobina Glucada/metabolismo , Perimenopausia/fisiología , Posmenopausia/fisiología , Premenopausia/fisiología , Adulto , Síntomas Afectivos/diagnóstico por imagen , Síntomas Afectivos/metabolismo , Síntomas Afectivos/fisiopatología , Amígdala del Cerebelo/diagnóstico por imagen , Corteza Cerebral/diagnóstico por imagen , Depresión/diagnóstico por imagen , Depresión/metabolismo , Depresión/fisiopatología , Femenino , Humanos , Imagen por Resonancia Magnética , Persona de Mediana Edad , Perimenopausia/metabolismo , Posmenopausia/metabolismo , Premenopausia/metabolismo
3.
Psychoneuroendocrinology ; 59: 25-36, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26010861

RESUMEN

The effects of postmenopausal hormone treatment on cognitive outcomes are inconsistent in the literature. Emerging evidence suggests that cognitive effects are influenced by specific hormone formulations, and that progesterone is more likely to be associated with positive outcomes than synthetic progestin. There are very few studies of unopposed progesterone in postmenopausal women, and none that use functional neuroimaging, a sensitive measure of neurobiological function. In this study of 29 recently postmenopausal women, we used functional MRI and neuropsychological measures to separately assess the effects of estrogen or progesterone treatment on visual and verbal cognitive function. Women were randomized to receive 90 days of either estradiol or progesterone counterbalanced with placebo. After each treatment arm, women were given a battery of verbal and visual cognitive function and working memory tests, and underwent functional MRI including verbal processing and visual working memory tasks. We found that both estradiol and progesterone were associated with changes in activation patterns during verbal processing. Compared to placebo, women receiving estradiol treatment had greater activation in the left prefrontal cortex, a region associated with verbal processing and encoding. Progesterone was associated with changes in regional brain activation patterns during a visual memory task, with greater activation in the left prefrontal cortex and right hippocampus compared to placebo. Both treatments were associated with a statistically non-significant increase in number of words remembered following the verbal task performed during the fMRI scanning session, while only progesterone was associated with improved neuropsychological measures of verbal working memory compared to placebo. These results point to potential cognitive benefits of both estrogen and progesterone.


Asunto(s)
Cognición/efectos de los fármacos , Estrógenos/farmacología , Posmenopausia/efectos de los fármacos , Progesterona/farmacología , Anciano , Terapia de Reemplazo de Estrógeno/métodos , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Memoria a Corto Plazo/efectos de los fármacos , Persona de Mediana Edad , Pruebas Neuropsicológicas , Placebos , Posmenopausia/metabolismo
4.
Fertil Steril ; 100(1): 200-7.e1, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23557757

RESUMEN

OBJECTIVE: To evaluate emotional processing in women with insulin-resistant polycystic ovary syndrome (IR-PCOS) and its relationship to glucose regulation and the mu-opioid system. DESIGN: Case-control pilot. SETTING: Tertiary referring medical center. PATIENT(S): Seven women with IR-PCOS and five non-insulin-resistant controls, aged 21-40 years, recruited from the general population. INTERVENTION(S): Sixteen weeks of metformin (1,500 mg/day) in women with IR-PCOS. MAIN OUTCOME MEASURE(S): Assessment of mood, metabolic function, and neuronal activation during an emotional task using functional magnetic resonance imaging (fMRI), and mu-opioid receptor availability using positive emission tomography (PET). RESULT(S): We found that insulin-resistant PCOS patients [1] had greater limbic activation during an emotion task than controls (n = 5); [2] trended toward decreased positive affect and increased trait anxiety; [3] after metformin treatment, had limbic activation that no longer differed from controls; and [4] had positive correlations between fMRI limbic activation during emotional processing and mu-opioid binding potential. CONCLUSION(S): Patients with IR-PCOS had greater regional activation during an emotion task than the controls, although this resolved with metformin therapy. Alterations in mu-opioid neurotransmission may underlie limbic system activity and mood disorders in IR-PCOS.


Asunto(s)
Emociones/fisiología , Neuroimagen Funcional/métodos , Resistencia a la Insulina/fisiología , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/psicología , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Estimulación Luminosa/métodos , Proyectos Piloto , Síndrome del Ovario Poliquístico/diagnóstico , Adulto Joven
5.
J Clin Endocrinol Metab ; 97(9): E1686-94, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22730514

RESUMEN

CONTEXT: Cognitive decline is prevalent in aging populations, and cognitive complaints are common during menopause. However, the extent of hormonal influence is unclear, particularly when considered independent of the aging process. OBJECTIVE: We sought to determine differences in cognitive function attributable to menopause, hypothesizing that differences would be associated with reproductive rather than chronological age. DESIGN AND SETTING: In this cross-sectional study at a university hospital, we combined neuropsychological measures with functional magnetic resonance imaging to comprehensively assess cognitive function. PARTICIPANTS: Sixty-seven menopausal women, aged 42-61 yr, recruited from a population-based menopause study, grouped into menopause stages based on hormonal and cycle criteria (premenopause, perimenopause, and postmenopause), participated in the study. MAIN OUTCOME MEASURES: Neuropsychological and functional magnetic resonance imaging measures of verbal, visual, and executive cognitive function. RESULTS: We found age-independent menopause effects on verbal function. Menopause groups differed in phonemic verbal fluency (F = 3.58, P < 0.019) and regional brain activation (inferior frontal cortex: corrected P < 0.000 right, P < 0.036 left; left prefrontal cortex: P < 0.012); left temporal pole: P < 0.001). Verbal measures correlated with estradiol and FSH (phonemic fluency: R = 0.249, P < 0.047 estradiol, R = -0.275, P < 0.029 FSH; semantic fluency: R = 0.318, P < 0.011 estradiol, R = -0.321, P < 0.010 FSH; right inferior frontal cortex: R = 0.364, P < 0.008 FSH; left inferior frontal cortex: R = -0.431, P < 0.001 estradiol, left prefrontal cortex: R = 0.279, P < 0.045 FSH; left temporal pole: R = -0.310, P < 0.024 estradiol, R = 0.451, P < 0.001 FSH; left parahippocampal gyrus: R = -0.278, P < 0.044 estradiol; left parietal cortex: R = -0.326, P < 0.017 estradiol). CONCLUSIONS: Results suggest that verbal fluency mechanisms are vulnerable during the menopausal transition. Targeted intervention may preserve function of this critical cognitive domain.


Asunto(s)
Envejecimiento/fisiología , Cognición/fisiología , Hormonas/fisiología , Menopausia/fisiología , Menopausia/psicología , Adulto , Encéfalo/fisiología , Estudios Transversales , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Humanos , Procesamiento de Imagen Asistido por Computador , Estudios Longitudinales , Imagen por Resonancia Magnética , Memoria/fisiología , Memoria a Corto Plazo/fisiología , Persona de Mediana Edad , Pruebas Neuropsicológicas , Perimenopausia/fisiología , Perimenopausia/psicología , Desempeño Psicomotor/fisiología , Globulina de Unión a Hormona Sexual/análisis , Testosterona/sangre , Percepción Visual/fisiología
6.
Behav Brain Res ; 226(1): 147-53, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21930160

RESUMEN

Despite considerable evidence for potential effects of estrogen on emotional processing, several studies of postmenopausal women who began hormone therapy (HT) remote from menopause report no effects of HT on emotional measures. As early HT initiation may preserve brain mechanisms, we examined effects of HT on emotional processing in postmenopausal women who started HT early after menopause. We performed a cross-sectional comparison of 52 postmenopausal women 66±5 years old, including 15 users of conjugated equine estrogen, 20 users of conjugated equine estrogen plus medroxyprogesterone acetate, and 17 who never used hormones (NT). All hormone users started therapy within two years of menopause, and received at least 10 years of continuous therapy. Outcomes were fMRI-detected brain activity and behavioral measures during an emotional processing picture rating task. During processing of positive pictures, NT women had greater activation than estrogen treated women in medial prefrontal cortex extending to the anterior cingulate, and more activation than estrogen plus progestin treated women in the insula. During processing of negative pictures, estrogen treated women had higher activation than NT women in the entorhinal cortex. Current compared to past HT users showed greater activation in the hippocampus and higher emotion recognition accuracy of neutral stimuli. Estrogen plus progestin treated women had slower response time than NT women when rating all pictures. In conclusion, hormone use was associated with differences in brain functional responses during emotional processing. These fMRI effects were more prominent than those observed for behavioral measures and involved brain regions implicated in cognitive-emotional integration.


Asunto(s)
Encéfalo/efectos de los fármacos , Emociones/efectos de los fármacos , Terapia de Reemplazo de Estrógeno/psicología , Estrógenos Conjugados (USP)/uso terapéutico , Acetato de Medroxiprogesterona/uso terapéutico , Posmenopausia/psicología , Anciano , Anciano de 80 o más Años , Mapeo Encefálico , Estrógenos Conjugados (USP)/administración & dosificación , Femenino , Humanos , Imagen por Resonancia Magnética , Acetato de Medroxiprogesterona/administración & dosificación , Persona de Mediana Edad , Neuroimagen , Tiempo de Reacción/efectos de los fármacos
7.
J Clin Endocrinol Metab ; 96(11): E1761-70, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21865354

RESUMEN

CONTEXT: The role of ovarian hormones in maintaining neuronal integrity and cognitive function is still debated. This study was undertaken to clarify the potential relationship between postmenopausal hormone use and the cholinergic system. OBJECTIVE: We hypothesized that early initiated hormone therapy (HT) preserves the cholinergic system and that estrogen therapy (ET) would be associated with higher levels of acetylcholinesterase activity in the posterior cingulate cortex and hippocampus compared to estrogen plus progestin therapy (EPT) or no HT. DESIGN AND SETTING: We conducted a cross-sectional study at a university teaching hospital. PATIENTS: Fifty postmenopausal women (age, 65.2 ± 0.7 yr) with early long-term HT (n = 34; 13 ET and 21 EPT) or no HT (n = 16) participated in the study. INTERVENTIONS: There were no interventions. MAIN OUTCOME MEASURE: We measured cholinergic activity (acetylcholinesterase) in the hippocampus and posterior cingulate brain regions as measured by N-[(11)C]methylpiperidin-4-yl propionate and positron emission tomography as a marker of cholinergic function. RESULTS: Significant effects of treatment on cholinergic activity measures were obtained in the left hippocampus (F = 3.56; P = 0.04), right hippocampus (F = 3.42; P = 0.04), and posterior cingulate (F = 3.76; P = 0.03). No significant effects were observed in a cortical control region. Post hoc testing identified greater cholinergic activity in the EPT group compared to the no-HT group in the left hippocampus (P = 0.048) and posterior cingulate (P = 0.045), with a nonstatistically significant trend in the right hippocampus (P = 0.073). CONCLUSIONS: A differential effect of postmenopausal ET and EPT on cholinergic neuronal integrity was identified in postmenopausal women. The findings are consistent with a preservation of cholinergic neuronal integrity in the EPT group.


Asunto(s)
Acetilcolinesterasa/metabolismo , Terapia de Reemplazo de Estrógeno/métodos , Estrógenos Conjugados (USP)/uso terapéutico , Estrógenos/uso terapéutico , Giro del Cíngulo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Menopausia/efectos de los fármacos , Factores de Edad , Anciano , Femenino , Giro del Cíngulo/enzimología , Hipocampo/enzimología , Humanos , Menopausia/metabolismo , Persona de Mediana Edad
8.
Fertil Steril ; 95(8): 2494-8, 2011 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-21486668

RESUMEN

This pilot study describes a relationship between insulin resistance and µ-opioid neurotransmission in limbic appetite and mood-regulating regions in women with polycystic ovary syndrome (PCOS), suggesting that insulin-opioid interactions may contribute to behavioral and reproductive pathologies of PCOS. We found that [1] patients with PCOS who are insulin-resistant (n = 7) had greater limbic µ-opioid receptor availability (nondisplaceable binding potential) than controls (n = 5); [2] receptor availability was correlated with severity of insulin resistance; and [3] receptor availability normalized after insulin-regulating treatment.


Asunto(s)
Encéfalo/metabolismo , Resistencia a la Insulina , Síndrome del Ovario Poliquístico/metabolismo , Receptores Opioides mu/metabolismo , betaendorfina/metabolismo , Adulto , Sitios de Unión , Encéfalo/efectos de los fármacos , Radioisótopos de Carbono , Estudios de Casos y Controles , Femenino , Fentanilo/análogos & derivados , Fentanilo/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Hipoglucemiantes/uso terapéutico , Imagen por Resonancia Magnética , Metformina/uso terapéutico , Michigan , Proyectos Piloto , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/fisiopatología , Tomografía de Emisión de Positrones , Receptores Opioides mu/efectos de los fármacos , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
9.
Menopause ; 17(4): 692-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20300040

RESUMEN

OBJECTIVE: The cognitive benefit of postmenopausal hormone use is controversial; however, timing of treatment close to menopause may increase the likelihood of preserving cognitive function. We examined the effects of early-initiation hormone use on visual working memory, hypothesizing that long-term hormone use is associated with greater brain activation during visual working memory. METHODS: This was a cross-sectional comparison of long-term early hormone users-current (n = 13) and past (n = 24; 2.1 +/- 1.0 years off hormones)-with never users (n = 18), using a visual memory task and functional magnetic resonance imaging (MRI). We evaluated 55 women older than 60 years at the University of Michigan's General Clinical Research Center. Hormone users had completed at least 10 continuous years of conjugated equine estrogens with or without medroxyprogesterone acetate, begun within 2 years of menopause. Women were excluded for illness, medication, intermittent estrogen use, phytoestrogen use, recent smoking, and MRI contraindications. The primary outcome was functional MRI-detected brain activity during the visual memory task. RESULTS: Compared with never users, both groups of hormone users had increased activation in the frontal and parietal cortices, insula, hippocampus, and cingulate; combined hormone users also had increased activation in the putamen and raphe (corrected P < 0.05 or uncorrected P < 0.001 with a priori hypothesis). Across the entire sample, the medial temporal cortex (P < 0.0001 right; P < 0.018 left) and right hippocampus (P < 0.0001) positively correlated with task performance. CONCLUSIONS: Hormone use was associated with increased brain activation during the visual memory task, in regions used for visual working memory. A positive correlation between activation and task performance suggests that early-initiation, long-term postmenopausal hormone use may benefit visual working memory.


Asunto(s)
Encéfalo/fisiología , Terapia de Reemplazo de Hormonas , Imagen por Resonancia Magnética , Memoria a Corto Plazo , Percepción Visual , Anciano , Estudios Transversales , Femenino , Humanos , Persona de Mediana Edad , Pruebas Neuropsicológicas , Posmenopausia
10.
Neurobiol Dis ; 30(3): 420-429, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18424057

RESUMEN

Clinical studies suggest that impaired glucose tolerance (IGT) is associated with the development of neuropathy. The aim of the current study was to determine if neuropathy developed in the female Zucker Diabetic Fatty (ZDF) rat, an animal model of IGT and type 2 diabetes. The ZDF rat develops impaired glucose tolerance (IGT) when fed a control diet, and frank diabetes when fed a high fat diet. Following 10 weeks of hyperglycemia, sensory nerve action potentials (SNAP) and compound motor action potentials (CMAP) were reduced and sensory conduction velocities were slowed (distal>proximal) in the tail and hind limb in ZDF animals with IGT and frank diabetes (p<0.01). Neuropathy was coupled with evidence of increased reactive oxygen species (ROS) and cellular injury in dorsal root ganglion (DRG) neurons from IGT animals. Our study supports the hypothesis that neuropathy develops in an animal model of IGT and is associated with evidence of oxidative injury in DRG and peripheral nerves.


Asunto(s)
Diabetes Mellitus/sangre , Neuropatías Diabéticas/sangre , Intolerancia a la Glucosa/sangre , Estrés Oxidativo/fisiología , Animales , Glucemia/genética , Glucemia/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Neuropatías Diabéticas/genética , Neuropatías Diabéticas/patología , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/patología , Ratas , Ratas Zucker
11.
Exp Neurol ; 211(2): 469-79, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18406405

RESUMEN

The mechanism/s leading to diabetic neuropathy are complex. Transforming growth factor-beta1 (TGF-beta1) has been associated with diabetic nephropathy and retinopathy but not neuropathy. In this study, changes in TGF-beta isoforms were examined in vivo and in vitro. Two groups of animals, streptozotocin diabetic with neuropathy and non-diabetic controls were examined at 4 weeks (n=10/group) and 12 weeks (n=8/group). In diabetic DRG using quantitative real-time PCR (QRT-PCR), TGF-beta1 and TGF-beta2 mRNA, but not TGF-beta3, was increased at 4 and 12 weeks. In sciatic nerve TGF-beta3 mRNA was primarily increased. Immunohistochemistry (DRG) and immunoblotting (sciatic nerve) showed similar differential protein expression. In sciatic nerve TGF-beta formed homo- and hetero-dimers, of which beta(2)/beta(3), beta(1)/beta(1), and beta(1)/beta(3) were significantly increased, while that of the TGF-beta(2)/beta(2) homodimer was decreased, in diabetic compared to non-diabetic rats. In vitro, pretreatment of embryonic DRG with TGF-beta neutralizing antibody prevents the increase in total TGF-beta protein observed with high glucose using immunoblotting. In high glucose conditions, combination with TGF-beta2>beta1 increases the percent of cleaved caspase-3 compared to high glucose alone and TGF-beta neutralizing antibody inhibits this increase. Furthermore, consistent with the findings in diabetic DRG and nerve, TGF-beta isoforms applied directly in vitro reduce neurite outgrowth, and this effect is partially reversed by TGF-beta neutralizing antibody. These findings implicate upregulation of TGF-beta in experimental diabetic peripheral neuropathy and indicate a novel mechanism of cellular injury related to elevated glucose levels. In combination, these findings indicate a potential new target for treatment of diabetic peripheral neuropathy.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/patología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Diabetes Mellitus Experimental/genética , Neuropatías Diabéticas/genética , Regulación de la Expresión Génica/fisiología , Masculino , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/fisiología , Factor de Crecimiento Transformador beta2/biosíntesis , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/fisiología , Factor de Crecimiento Transformador beta3/biosíntesis , Factor de Crecimiento Transformador beta3/genética , Factor de Crecimiento Transformador beta3/fisiología
12.
Curr Drug Targets ; 9(1): 85-93, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18220716

RESUMEN

Multiple in vivo and in vitro studies show that excessive release of glutamate, and subsequent activation of ionotropic glutamate receptors (iGluRs) and some metabotropic glutamate receptors (mGluRs) cause neuronal cell death through either necrosis or apoptosis. However, recently alternative evidence has shown that mGluRs have modulatory effects on excitotoxicity and neuronal cell death. Metabotropic glutamate receptors form a family of eight subtypes (mGluR1-8), subdivided into three groups (I-III) that initiate their biological effects by G protein-linked intracellular signal transduction. Their expression throughout the mammalian nervous system implicates these receptors as essential mediators of a cell's fate during injury to the nervous system. Activation of group-II (mGluR2 and -3) or group-III metabotropic glutamate receptors (mGluR4, -6, -7 and -8) has been established to be neuroprotective in vitro and in vivo. In contrast, group-I mGluRs (mGluR1 and -5) need to be antagonized in order to evoke protection. The pathological signaling pathways associated with diabetic neuropathy are complex and this influences development of appropriate therapies. The Group II mGluRs target several signaling pathways affected in diabetic neuropathy, prevent cellular injury in the peripheral nervous system, and may provide a novel mechanism for treatment of diabetic neuropathy. Direct or indirect activation of mGluR2/3 in animal models protects against development of diabetic neuropathy. The potential mechanisms and role of mGluRs in protection against diabetic neuropathy will be reviewed.


Asunto(s)
Neuropatías Diabéticas/tratamiento farmacológico , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Animales , Neuropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo , Receptores de Glutamato Metabotrópico/metabolismo , Transducción de Señal
13.
J Neurochem ; 101(2): 342-54, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17402968

RESUMEN

High glucose concentrations cause oxidative injury and programmed cell death in neurons, and can lead to diabetic neuropathy. Activating the type 3 metabotropic glutamate receptor (mGluR3) prevents glucose-induced oxidative injury in dorsal root ganglion neurons co-cultured with Schwann cells. To determine the mechanisms of protection, studies were performed in rat dorsal root ganglion neuron-Schwann cell co-cultures. The mGluR3 agonist 2R,4R-4-aminopyrrolidine-2,4-dicarboxylate prevented glucose-induced inner mitochondrial membrane depolarization, reactive oxygen species accumulation, and programmed cell death, and increased glutathione (GSH) concentration in co-cultured neurons and Schwann cells, but not in neurons cultured without Schwann cells. Protection was diminished in neurons treated with the GSH synthesis inhibitor l-buthionine-sulfoximine, suggesting that mGluR-mediated protection requires GSH synthesis. GSH precursors and the GSH precursor GSH-ethyl ester also protected neurons from glucose-induced injury, indicating that GSH synthesis in Schwann cells, and transport of reaction precursors to neurons, may underlie mGluR-mediated neuroprotection. These results support the conclusions that activating glial mGluR3 protects neurons from glucose-induced oxidative injury by increasing free radical scavenging and stabilizing mitochondrial function, through increased GSH antioxidant defense.


Asunto(s)
Ganglios Espinales/metabolismo , Glucosa/toxicidad , Glutatión/metabolismo , Neuronas Aferentes/metabolismo , Estrés Oxidativo/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Antimetabolitos/toxicidad , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Butionina Sulfoximina/toxicidad , Células Cultivadas , Técnicas de Cocultivo , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/fisiopatología , Agonistas de Aminoácidos Excitadores/farmacología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiopatología , Glucosa/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Espacio Intracelular/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Neuronas Aferentes/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
14.
J Neurochem ; 89(1): 90-9, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15030392

RESUMEN

Glutamate carboxypeptidase II (GCP II) inhibition has previously been shown to be protective against long-term neuropathy in diabetic animals. In the current study, we have determined that the GCP II inhibitor 2-(phosphonomethyl) pentanedioic acid (2-PMPA) is protective against glucose-induced programmed cell death (PCD) and neurite degeneration in dorsal root ganglion (DRG) neurons in a cell culture model of diabetic neuropathy. In this model, inhibition of caspase activation is mediated through the group II metabotropic glutamate receptor, mGluR3. 2-PMPA neuroprotection is completely reversed by the mGluR3 antagonist (S)-alpha-ethylglutamic acid (EGLU). In contrast, group I and III mGluR inhibitors have no effect on 2-PMPA neuroprotection. Furthermore, we show that two mGluR3 agonists, the direct agonist (2R,4R)-4-aminopyrrolidine-2, 4-dicarboxylate (APDC) and N-acetyl-aspartyl-glutamate (NAAG) provide protection to neurons exposed to high glucose conditions, consistent with the concept that 2-PMPA neuroprotection is mediated by increased NAAG activity. Inhibition of GCP II or mGluR3 may represent a novel mechanism to treat neuronal degeneration under high-glucose conditions.


Asunto(s)
Dipéptidos/farmacología , Glucosa/toxicidad , Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Neuropatías Diabéticas/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Ganglios Espinales/citología , Glutamato Carboxipeptidasa II/metabolismo , Neuritas/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Compuestos Organofosforados/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Células de Schwann/citología , Células de Schwann/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...