Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 369
Filtrar
1.
Sci Rep ; 11(1): 20134, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34635736

RESUMEN

DNA methylation shifts in Hypothalamic-pituitary-adrenal (HPA) axis related genes is reported in psychiatric disorders including hypersexual disorder. This study, comprising 20 dexamethasone suppression test (DST) non-suppressors and 73 controls, examined the association between the HPA axis dysregulation, shifts in DNA methylation of HPA axis related genes and importantly, gene expression. Individuals with cortisol level ≥ 138 nmol/l, after the low dose (0.5 mg) dexamethasone suppression test (DST) were classified as non-suppressors. Genome-wide methylation pattern, measured in whole blood using the EPIC BeadChip, investigated CpG sites located within 2000 bp of the transcriptional start site of key HPA axis genes, i.e.: CRH, CRHBP, CRHR-1, CRHR-2, FKBP5 and NR3C1. Regression models including DNA methylation M-values and the binary outcome (DST non-suppression status) were performed. Gene transcripts with an abundance of differentially methylated CpG sites were identified with binomial tests. Pearson correlations and robust linear regressions were performed between CpG methylation and gene expression in two independent cohorts. Six of 76 CpG sites were significantly hypermethylated in DST non-suppressors (nominal P < 0.05), associated with genes CRH, CRHR1, CRHR2, FKBP5 and NR3C1. NR3C1 transcript AJ877169 showed statistically significant abundance of probes differentially methylated by DST non-suppression status and correlated with DST cortisol levels. Further, methylation levels of cg07733851 and cg27122725 were positively correlated with gene expression levels of the NR3C1 gene. Methylation levels of cg08636224 (FKBP5) correlated with baseline cortisol and gene expression. Our findings revealed that DNA methylation shifts are involved in the altered mechanism of the HPA axis suggesting that new epigenetic targets should be considered behind psychiatric disorders.


Asunto(s)
Metilación de ADN , Dexametasona/antagonistas & inhibidores , Regulación de la Expresión Génica , Sistema Hipotálamo-Hipofisario/patología , Trastornos Parafílicos/patología , Sistema Hipófiso-Suprarrenal/patología , Disfunciones Sexuales Psicológicas/patología , Adolescente , Adulto , Anciano , Biomarcadores/análisis , Estudios de Casos y Controles , Dexametasona/administración & dosificación , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Humanos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Persona de Mediana Edad , Trastornos Parafílicos/genética , Trastornos Parafílicos/metabolismo , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Disfunciones Sexuales Psicológicas/genética , Adulto Joven
2.
J Endocrinol Invest ; 44(3): 609-619, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32686043

RESUMEN

PURPOSE: Data on endothelial derangements in patients with non-functioning adrenal incidentaloma (NFAI) are scarce. METHODS: We investigated if NFAI patients present clinical, biochemical and endothelial alterations compared to individuals without an adrenal lesion and also the associations among these variables. Forty-two NFAI and 40 controls were evaluated. NFAI diagnosis and controls were defined according to the current guidelines and based on a normal adrenal imaging exam, respectively. Body composition was evaluated by dual emission X-ray absorptiometry. Endothelial reactivity was assessed by two methods: tonometry (Endo-PAT®) and laser speckle contrast imaging (LSCI). RESULTS: There were no differences between groups regarding age, gender, ethnicity, smoking status, and statin use. The frequency of metabolic syndrome according to the International Diabetes Federation criteria was 69% and 57.9%, respectively in NFAI and controls (p = 0.36), whereas the atherosclerotic cardiovascular disease (ASCVD) risk was 63.4% and 66.7% (p = 0.81). The clinical, laboratory, and anthropometric characteristics, as well as body composition, were similar between the groups. Additionally, any differences between groups were observed on endothelial reactivity tests. Nevertheless, we noted an association between cortisol levels after 1 mg-dexamethosone suppression test (1 mg-DST) and the duration of post-occlusive reactive hyperemia tested on microcirculation (r = 0.30; p = 0.03). NFAI patients require more antihypertensive drugs to achieve blood pressure control (p = 0.04). The number of antihypertensive drugs used to control blood pressure correlated with cortisol levels after 1 mg-DST (r = 0.29; p = 0.03). CONCLUSIONS: Since both groups herein investigated had a high frequency of metabolic syndrome and ASCVD risk, it might explain similarities observed on endothelial reactivity. Nevertheless, prolonged reactive hyperemia response on microcirculation was correlated with cortisol levels under suppression.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/complicaciones , Biomarcadores/sangre , Enfermedades Cardiovasculares/diagnóstico , Dexametasona/antagonistas & inhibidores , Hidrocortisona/sangre , Hiperemia/diagnóstico , Síndrome Metabólico/diagnóstico , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/etiología , Estudios de Casos y Controles , Estudios Transversales , Femenino , Estudios de Seguimiento , Humanos , Hiperemia/sangre , Hiperemia/etiología , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/etiología , Persona de Mediana Edad , Pronóstico
3.
Drug Des Devel Ther ; 14: 4451-4463, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33122889

RESUMEN

PURPOSE: Glucocorticoids are used for the treatment of inflammatory diseases, but glucocorticoid treatment is associated with bone damage. Resveratrol is a phytoalexin found in many plants, and we investigated its protective role on dexamethasone-induced dysfunction in MC3T3-E1 cells and primary osteoblasts. MATERIALS AND METHODS: MC3T3-E1 cells and primary osteoblasts were treated with dexamethasone in the presence/absence of different doses of resveratrol for 24 or 48 h. Then, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium (MTT) and lactate dehydrogenase (LDH) assays were used to evaluate cell viability. Apoptosis was analyzed by a flow cytometry. An alkaline phosphatase (ALP) activity assay and Alizarin Red S staining were used to study osteoblast differentiation. Expression of osteoblast-related genes was measured by real-time reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The AMP-activated protein kinase (AMPK) signaling pathway and mitochondrial expression of superoxide dismutase were evaluated by Western blotting. Intracellular reactive oxygen species (ROS), adenosine triphosphate (ATP) content, mitochondrial-complex activity, and mitochondrial DNA content were measured to evaluate mitochondrial function. RESULTS: Resveratrol induced the proliferation and inhibited apoptosis of osteoblasts in the presence of dexamethasone. Resveratrol increased the ALP activity and mineralization of osteoblasts. Resveratrol also attenuated dexamethasone-induced inhibition of mRNA expression of osteogenesis maker genes, including bone morphogenetic protein-2, osteoprotegerin, runt-related transcription factor-2, and bone Gla protein. Resveratrol alleviated dexamethasone-induced mitochondrial dysfunction. Resveratrol strongly stimulated expression of peroxisome proliferator-activated receptor-γ coactivator 1α and sirtuin-3 genes, as well as their downstream target gene superoxide dismutase-2. Resveratrol induced phosphorylation of AMPK and acetyl-CoA carboxylase (ACC). Blockade of AMPK signaling using compound C reversed the protective effects of resveratrol against dexamethasone. CONCLUSION: Resveratrol showed protective effects against dexamethasone-induced dysfunction of osteoblasts by activating AMPK signaling.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Dexametasona/antagonistas & inhibidores , Osteoblastos/efectos de los fármacos , Sustancias Protectoras/farmacología , Resveratrol/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Osteoblastos/metabolismo , Fosforilación/efectos de los fármacos , Relación Estructura-Actividad
4.
Eur Rev Med Pharmacol Sci ; 24(10): 5691-5696, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32495904

RESUMEN

OBJECTIVE: To elucidate the role of Prunella vulgaris L (PVL) in protecting glucocorticoids (GC)-induced osteogenesis inhibition, thereafter, protecting the deterioration of osteoporosis (OP). MATERIALS AND METHODS: Cell Counting Kit-8 (CCK-8) assay was conducted to assess the influence of PVL treatment on MSCs viability. Osteogenesis in MSCs was induced by Dexamethasone (DEX) stimulation. Regulatory effects of PVL on osteogenesis-related gene expressions, ALP activity, and mineralization ability in DEX-induced MSCs were determined. At last, protein levels of p-Smad1/5/9 and total-Smad1/5/9 influenced by DEX and PVL were measured by Western blot. RESULTS: PVL treatment did not pose a time- or dose-dependent influence on MSCs viability. DEX induction in MSCs downregulated ALP, RUNX2, Bglap, and Osterix. ALP activity and mineralization in DEX-induced MSCs were suppressed. Downregulated osteogenesis-related genes decreased ALP activity and mineralization in MSCs undergoing DEX stimulation were partially reversed by PVL treatment. Moreover, the downregulated p-Smad1/5/9 level in DEX-induced MSCs was elevated by PVL treatment, while total-Smad1/5/9 was not affected. CONCLUSIONS: PVL alleviated GC-induced suppression in MSCs osteogenesis by activating the Smad pathway, thereafter, protecting the deterioration of OP.


Asunto(s)
Dexametasona/antagonistas & inhibidores , Glucocorticoides/antagonistas & inhibidores , Células Madre Mesenquimatosas/efectos de los fármacos , Sustancias Protectoras/farmacología , Prunella/química , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Células Cultivadas , Dexametasona/farmacología , Glucocorticoides/farmacología , Humanos , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Sustancias Protectoras/química , Proteína Smad1/antagonistas & inhibidores , Proteína Smad5/antagonistas & inhibidores , Proteína Smad8/antagonistas & inhibidores
5.
Drug Des Devel Ther ; 14: 195-206, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32021104

RESUMEN

BACKGROUND: Osteoporosis is a chronic bone metabolism disorder affecting millions of the world population. The RANKL/RANK/OPG signaling pathway has been confirmed to be the main regulator of osteoporosis. It is of great interest to identify appropriate therapeutic agents that can regulate the RANKL/RANK/OPG pathway. Baicalin (BA) is a well-known traditional Chinese medicine formula against various inflammatory diseases with a proven role of the RANKL/RANK/OPG pathway regulation. However, the potential effect of BA on osteoporosis and the mechanisms underlying this remain unclear. In the present study, we aimed to evaluate the efficacy of BA in the prevention of dexamethasone (DEX)-induced osteoporosis in zebrafish. METHODS: In this study, growth and development changes of zebrafish and calcein staining were assessed with a micrograph. The expression levels of RANKL and OPG and transcription factors in response to DEX induction and BA administration were evaluated by Western blotting and qRT-PCR. In addition, the intermolecular interactions of BA and RANKL were investigated by molecular docking. RESULTS: Results show that BA enhances the growth and development of dexamethasone (DEX)-induced osteoporosis in zebrafish larvae. Calcein staining and calcium and phosphorus determination revealed that BA ameliorates mineralization of DEX-induced osteoporosis zebrafish larvae. BA also regulates the expression of RANKL and OPG and hampers the changes in gene expression related to bone formation and resorption under the induction of DEX in zebrafish. It can be inferred by molecular docking that BA may interact directly with the extracellular domain of RANKL. CONCLUSION: The findings, herein, reveal that BA ameliorates DEX-induced osteoporosis by regulation of the RANK/RANKL/OPG signaling pathway.


Asunto(s)
Dexametasona/antagonistas & inhibidores , Flavonoides/farmacología , Osteoporosis/tratamiento farmacológico , Osteoprotegerina/metabolismo , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Flavonoides/administración & dosificación , Larva/efectos de los fármacos , Larva/metabolismo , Simulación del Acoplamiento Molecular , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoporosis/inducido químicamente , Osteoprotegerina/genética , Ligando RANK/genética , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Pez Cebra
6.
Brain Res Bull ; 152: 225-235, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31357009

RESUMEN

Calbindin-D9k (CaBP-9k), one of the major calcium-binding and calcium-buffering proteins, is important in the physiological functioning of organs. The neuroanatomical localization of CaBP-9k in the rodent brain has not been reported; thus, this study investigated the neuroanatomical distribution of CaBP-9k and the regulation of CaBP-9k expression on steroid hormones in the immature rat brain. To confirm the influence of steroid hormones on CaBP-9k expression, immature female rats were injected for 5 days with estrogen (E2), progesterone (P4), dexamethasone (DEX), and their antagonists (ICI 182, 780 and RU 486). The localization and expression of the CaBP-9k protein in brain regions were identified by immunofluorescence and western blot assays, respectively. We observed that CaBP-9k expression was especially strong in hypothalamus, cerebellum, and brain stem. In addition, CaBP-9k was colocalized with mature-, GABAergic, dopaminergic, and oxytocinergic neurons. We also observed that the CaBP-9k protein level was significantly increased by P4 and reversed by antagonist RU 486 treatment in immature rat brain. In summary, CaBP-9k positive cells have a wide distribution in the immature rat brain, and CaBP-9k expression is regulated by P4. We suggest that CaBP-9k expression regulated by steroid hormone may serve as an important regulator of cytosolic calcium concentration in the brain.


Asunto(s)
Proteína G de Unión al Calcio S100/metabolismo , Animales , Western Blotting , Encéfalo/metabolismo , Calbindinas/metabolismo , Dexametasona/antagonistas & inhibidores , Dexametasona/farmacología , Estradiol/farmacología , Antagonistas de Estrógenos , Femenino , Mifepristona/farmacología , Progesterona/antagonistas & inhibidores , Progesterona/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Arch Biochem Biophys ; 664: 157-166, 2019 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-30771297

RESUMEN

Prevention of muscle wasting is known to contribute to improving the quality of life and extending a healthy life. Recently, we have reported that licorice flavonoid oil containing glabridin, which is a prenylated isoflavone, enhances muscle mass in mice. In this study, we investigated the prevention effect of glabridin on dexamethasone-induced muscle atrophy and clarified its mechanism in cultured myotubes and in muscle of mice. Treatment with glabridin to C2C12 myotubes inhibited dexamethasone-induced protein degradation through dexamethasone-induced expression of ubiquitin ligases, MuRF1 and Cbl-b, but not atrogin-1. Mechanistically, glabridin inhibited nuclear translocation of the glucocorticoid receptor. Glabridin directly bound to the glucocorticoid receptor, resulting in the inhibition of binding between dexamethasone and the receptor protein. Glabridin also inhibited dexamethasone-induced phosphorylation of p38 and FoxO3a, as the upstream for the induction of ubiquitin ligases in C2C12 myotubes. Moreover, the glabridin-induced inhibition of protein degradation was eliminated by knockdown of the glucocorticoid receptor, but not by p38 knockdown. These data indicated that the inhibitory mechanism of glabridin against dexamethasone-induced muscle atrophy was mainly mediated by the inhibition of binding between dexamethasone and the glucocorticoid receptor in myotubes. Oral administration of glabridin prevented dexamethasone-induced protein degradation in the tibialis anterior muscle of mice. It was confirmed that glabridin inhibited dexamethasone-induced nuclear translocation of the glucocorticoid receptor and phosphorylation of FoxO3a in the muscle of mice. These findings suggest that glabridin is an effective food ingredient for the prevention of glucocorticoid-induced skeletal muscle atrophy.


Asunto(s)
Dexametasona/antagonistas & inhibidores , Isoflavonas/farmacología , Atrofia Muscular/prevención & control , Fenoles/farmacología , Animales , Línea Celular , Dexametasona/metabolismo , Dexametasona/farmacología , Proteína Forkhead Box O3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Atrofia Muscular/inducido químicamente , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Biosci Biotechnol Biochem ; 83(4): 659-665, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30543144

RESUMEN

In the present study, we examined the effects of fluvoxamine on nerve growth factor (NGF)-induced neurite outgrowth inhibition by dexamethasone (DEX) in PC12 cells. Fluvoxamine increased NGF-induced neurite outgrowth. Compared with co-treatment with NGF and fluvoxamine, p-Akt levels were higher than the values without fluvoxamine. The phosphorylated extracellular regulated kinase 1/2 levels were slightly increased by co-treatment with NGF and fluvoxamine. Fluvoxamine concentration-dependently improved NGF-induced neurite outgrowth inhibition by DEX. Fluvoxamine also improved the decrease in the NGF-induced p-Akt level caused by DEX. Interestingly, the sigma-1 receptor antagonist NE-100 blocked the improvement effects of fluvoxamine on NGF-induced neurite outgrowth inhibition by DEX. The selective sigma-1 receptor agonist PRE-084 also improved NGF-induced neurite outgrowth inhibition by DEX, which is blocked by NE-100. These results indicate that the improvement effects of fluvoxamine on NGF-induced neurite outgrowth inhibition by DEX may be attributable to the phosphorylation of Akt and the sigma-1 receptor.


Asunto(s)
Ansiolíticos/farmacología , Fluvoxamina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Animales , Anisoles/farmacología , Diferenciación Celular/efectos de los fármacos , Dexametasona/antagonistas & inhibidores , Dexametasona/farmacología , Glucocorticoides/antagonistas & inhibidores , Glucocorticoides/farmacología , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfolinas/farmacología , Factor de Crecimiento Nervioso/farmacología , Proyección Neuronal/genética , Neuronas/citología , Neuronas/metabolismo , Células PC12 , Fosforilación/efectos de los fármacos , Propilaminas/farmacología , Proteínas Proto-Oncogénicas c-akt/agonistas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptores sigma/agonistas , Receptores sigma/antagonistas & inhibidores , Receptores sigma/genética , Receptores sigma/metabolismo , Transducción de Señal , Receptor Sigma-1
9.
J Nat Prod ; 80(5): 1505-1513, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28489375

RESUMEN

Species of the genus Laserpitium have been used traditionally to treat inflammation and infection. From the herb of Laserpitium zernyi, six new compounds were isolated and their structures elucidated (using IR, NMR, HRMS data) as derivatives of 8-daucene-2,4,10-triol (1, 2, and 4), 7-daucene-2,4,10-triol (3), a lapiferin derivative featuring a C-2 ester moiety (5), and a daucane featuring an exomethylene group at C-8 (6). Also isolated were the rare daucanes vaginatin (7) and laserpitin (8). In a search for selective glucocorticoid receptor (GR) modulators, the compounds were tested for their capacity to inhibit NF-κB and AP-1 pro-inflammatory factors and for a potential competitive effect on a dexamethasone (Dex)-induced GR-driven glucocorticoid response element (GRE) reporter gene. The new 2ß-angeloyloxy-10α-acetoxy-8-daucene-2,4,10-triol (2) significantly inhibited transactivation of both NF-κB and AP-1, while vaginatin (7) was the most active of the compounds tested in blocking AP-1. Both compounds competitively repressed Dex-induced GRE-driven promoter activities, indicative of a potential role for GR. In addition, a decreased potential to inhibit NF-κB was apparent in GR knockout A549 cells. In line with the transcriptional assays, compounds 2 and 7 also significantly lowered CCL-2 chemokine production, albeit to a lesser extent than Dex. The results suggest that daucanes may be interesting candidates in the search for compounds with GR-modulating activities.


Asunto(s)
Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacología , Apiaceae/química , Compuestos Bicíclicos con Puentes/aislamiento & purificación , Compuestos Bicíclicos con Puentes/farmacología , Dexametasona/antagonistas & inhibidores , Dexametasona/química , FN-kappa B/antagonistas & inhibidores , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/química , Sesquiterpenos/aislamiento & purificación , Sesquiterpenos/farmacología , Antiinflamatorios/química , Compuestos Bicíclicos con Puentes/química , Ésteres , Humanos , Espectroscopía de Resonancia Magnética , Estructura Molecular , FN-kappa B/química , Sesquiterpenos/química , Factor de Transcripción AP-1 , Activación Transcripcional
10.
Arch Toxicol ; 91(1): 231-246, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27052459

RESUMEN

Astrocytes, the largest cell population in the human brain, are powerful inflammatory effectors. Several studies have examined the interaction of activated astrocytes with neurons, but little is known yet about human neurotoxicity under such situations and about strategies of neuronal rescue. To address this question, immortalized murine astrocytes (IMA) were combined with human LUHMES neurons and stimulated with an inflammatory (TNF, IL-1) cytokine mix (CM). Neurotoxicity was studied both in co-cultures and in monocultures after transfer of conditioned medium from activated IMA. Interventions with >20 drugs were used to profile the model system. Control IMA supported neurons and protected them from neurotoxicants. Inflammatory activation reduced this protection, and prolonged exposure of co-cultures to CM triggered neurotoxicity. Neither the added cytokines nor the release of NO from astrocytes were involved in this neurodegeneration. The neurotoxicity-mediating effect of IMA was faithfully reproduced by human astrocytes. Moreover, glia-dependent toxicity was also observed, when IMA cultures were stimulated with CM, and the culture medium was transferred to neurons. Such neurotoxicity was prevented when astrocytes were treated by p38 kinase inhibitors or dexamethasone, whereas such compounds had no effect when added to neurons. Conversely, treatment of neurons with five different drugs, including resveratrol and CEP1347, prevented toxicity of astrocyte supernatants. Thus, the sequential IMA-LUHMES neuroinflammation model is suitable for separate profiling of both glial-directed and directly neuroprotective strategies. Moreover, direct evaluation in co-cultures of the same cells allows for testing of therapeutic effectiveness in more complex settings, in which astrocytes affect pharmacological properties of neurons.


Asunto(s)
Astrocitos/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Citocinas/agonistas , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Neurotoxinas/toxicidad , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Astrocitos/citología , Astrocitos/inmunología , Astrocitos/metabolismo , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Dexametasona/antagonistas & inhibidores , Dexametasona/farmacología , Antagonistas de Hormonas/farmacología , Humanos , Interleucina-1beta/agonistas , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Mifepristona/farmacología , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/citología , Neuronas/inmunología , Neuronas/metabolismo , Neurotoxinas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Factor de Necrosis Tumoral alfa/agonistas , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
11.
Cell Death Dis ; 7: e2235, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27228350

RESUMEN

Long-term use of glucocorticoids is a widespread clinical problem, which currently has no effective solution other than discontinuing the use. Eicosapentaenoic acid (EPA), an omega-3 long chain polyunsaturated fatty acid (n-3 PUFA), which is largely contained in fish or fish oil, has been reported to promote cell viability and improve bone metabolism. However, little is known about the effects of EPA on dexamethasome (Dex)-induced cell apoptosis. In this study, we showed that EPA-induced autophagy of murine bone marrow-derived mesenchymal stem cells (mBMMSCs). Meanwhile, EPA, but not arachidonic acid (AA), markedly inhibited Dex-induced apoptosis and promoted the viability of mBMMSCs. We also observed that EPA-induced autophagy was modulated by GPR120, but not GPR40. Further experiments showed that the mechanism of EPA-induced autophagy associated with GPR120 modulation involved an increase in the active form of AMP-activated protein kinase and a decrease in the activity of mammalian target of RAPA. The protective effect of EPA on Dex-induced apoptosis via GPR120-meditated induction of adaptive autophagy was supported by in vivo experiments. In summary, our findings may have important implications in developing future strategies to use EPA in the prevention and therapy of the side effects induced by long-term Dex-abuse.


Asunto(s)
Autofagia/efectos de los fármacos , Dexametasona/antagonistas & inhibidores , Ácido Eicosapentaenoico/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ácido Araquidónico/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Supervivencia Celular/efectos de los fármacos , Dexametasona/farmacología , Femenino , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Cultivo Primario de Células , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
12.
J Physiol Biochem ; 72(2): 245-53, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26944908

RESUMEN

We previously observed that sarcoendoplasmic reticulum Ca(2+) ATPase (SERCA) blockade by cyclopiazonic acid (CPA) significantly potentiates serotonin (5-hydroxytryptamine (5-HT))-induced vascular contractions. Furthermore, 5-HT receptor antagonist methysergide partially inhibited CPA-potentiated 5-HT contractions. In the present study, we further investigated whether SERCA inhibition potentiates 5-HT-induced Ca(2+) responses along with attenuating the receptor antagonism by store-operated Ca(2+) (SOC) entry and protein kinase C (PKC)-mediated mechanisms. The effects of dexamethasone that was previously shown to induce SOC entry and enhance 5-HT responses were also tested. For this purpose, intracellular Ca(2+) levels were monitored in A7r5 embryonic rat vascular smooth muscle cells by spectrofluorometry using the fluorescent indicator fura-2. The results showed that CPA, although not dexamethasone, significantly potentiated 5-HT-induced Ca(2+) elevations. Ketanserin partially decreased 5-HT-induced and CPA-potentiated Ca(2+) elevations whereas both PKC inhibitor D-sphingosine and SOC entry blocker 2-aminoethoxydiphenyl borate (2-APB) abolished the remaining responses. The data suggests that diminished antagonistic effect on 5-HT-induced Ca(2+) elevations in the presence of SERCA inhibition is induced by SOC entry and PKC activation.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Músculo Liso Vascular/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , Agonistas de Receptores de Serotonina/farmacología , Serotonina/metabolismo , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Línea Celular , Dexametasona/antagonistas & inhibidores , Dexametasona/farmacología , Inhibidores Enzimáticos/química , Indoles/antagonistas & inhibidores , Ketanserina/farmacología , Metisergida/farmacología , Músculo Liso Vascular/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ratas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Serotonina/química , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/química , Vasoconstrictores/antagonistas & inhibidores , Vasoconstrictores/farmacología , Vasodilatadores/antagonistas & inhibidores , Vasodilatadores/farmacología
13.
Biochem Biophys Res Commun ; 471(4): 545-52, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26891866

RESUMEN

Excessive glucocorticoid (GC) usage may lead to non-traumatic femoral head osteonecrosis. Dexamethasone (Dex) exerts cytotoxic effect to cultured osteoblasts. Here, we investigated the potential activity of Compound 13 (C13), a novel α1 selective AMP-activated protein kinase (AMPK) activator, against the process. Our data revealed that C13 pretreatment significantly attenuated Dex-induced apoptosis and necrosis in both osteoblastic-like MC3T3-E1 cells and primary murine osteoblasts. AMPK activation mediated C13' cytoprotective effect in osteoblasts. The AMPK inhibitor Compound C, shRNA-mediated knockdown of AMPKα1, or dominant negative mutation of AMPKα1 (T172A) almost abolished C13-induced AMPK activation and its pro-survival effect in osteoblasts. On the other hand, forced AMPK activation by adding AMPK activator A-769662 or exogenous expression a constitutively-active (ca) AMPKα1 (T172D) mimicked C13's actions and inhibited Dex-induced osteoblast cell death. Meanwhile, A-769662 or ca-AMPKα1 almost nullified C13's activity in osteoblast. Further studies showed that C13 activated AMPK-dependent nicotinamide adenine dinucleotide phosphate (NADPH) pathway to inhibit Dex-induced reactive oxygen species (ROS) production in MC3T3-E1 cells and primary murine osteoblasts. Such effects by C13 were almost reversed by Compound C or AMPKα1 depletion/mutation. Together, these results suggest that C13 alleviates Dex-induced osteoblast cell death via activating AMPK signaling pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Apoptosis/efectos de los fármacos , Dexametasona/efectos adversos , Glucocorticoides/efectos adversos , Osteoblastos/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/genética , Animales , Apoptosis/genética , Compuestos de Bifenilo , Línea Celular , Dexametasona/antagonistas & inhibidores , Activación Enzimática , Glucocorticoides/antagonistas & inhibidores , Ratones , Mutación , NADP/metabolismo , Osteoblastos/enzimología , Cultivo Primario de Células , Pironas/farmacología , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Tiofenos/farmacología
14.
J Nutr ; 145(10): 2300-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26338887

RESUMEN

BACKGROUND: Long-term dietary curcumin (>12 wk) improves metabolic homeostasis in obese mice by sensitizing insulin signaling and reducing hepatic gluconeogenesis. Whether these occur only secondary to its chronic anti-inflammatory and antioxidative functions is unknown. OBJECTIVE: In this study, we assessed the insulin sensitization effect of short-term curcumin gavage in a rapid dexamethasone-induced insulin resistance mouse model, in which the chronic anti-inflammatory function is eliminated. METHODS: Six-week-old male C57BL/6 mice received an intraperitoneal injection of dexamethasone (100 mg/kg body weight) or phosphate-buffered saline every day for 5 d, with or without simultaneous curcumin gavage (500 mg/kg body weight). On day 7, insulin tolerance tests were performed. After a booster dexamethasone injection and curcumin gavage on day 8, blood glucose and insulin concentrations were measured. Liver tissues were collected on day 10 for quantitative polymerase chain reaction and Western blotting to assess gluconeogenic gene expression, insulin signaling, and the expression of fibroblast growth factor 21 (FGF21). Primary hepatocytes from separate, untreated C57BL/6 mice were used for testing the in vitro effect of curcumin treatment. RESULTS: Dexamethasone injection impaired insulin tolerance (P < 0.05) and elevated ambient plasma insulin concentrations by ~2.7-fold (P < 0.01). Concomitant curcumin administration improved insulin sensitivity and reduced hepatic gluconeogenic gene expression. The insulin sensitization effect of curcumin was demonstrated by increased stimulation of S473 phosphorylation of protein kinase B (P < 0.01) in the dexamethasone-treated mouse liver, as well as the repression of glucose production in primary hepatocytes (P < 0.001). Finally, curcumin gavage increased FGF21 expression by 2.1-fold in the mouse liver (P < 0.05) and curcumin treatment increased FGF21 expression in primary hepatocytes. CONCLUSION: These observations suggest that the early beneficial effect of curcumin intervention in dexamethasone-treated mice is the sensitization of insulin signaling, involving the stimulation of FGF21 production, a known insulin sensitizer.


Asunto(s)
Antioxidantes/uso terapéutico , Curcumina/uso terapéutico , Suplementos Dietéticos , Factores de Crecimiento de Fibroblastos/agonistas , Resistencia a la Insulina , Hígado/metabolismo , Estado Prediabético/prevención & control , Animales , Antioxidantes/metabolismo , Glucemia/análisis , Células Cultivadas , Curcumina/metabolismo , Dexametasona/antagonistas & inhibidores , Dexametasona/toxicidad , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucocorticoides/antagonistas & inhibidores , Glucocorticoides/toxicidad , Gluconeogénesis/efectos de los fármacos , Células Hep G2 , Humanos , Insulina/sangre , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Estado Prediabético/inducido químicamente , Estado Prediabético/metabolismo , Estado Prediabético/patología , Distribución Aleatoria , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Genet Mol Res ; 14(2): 6968-77, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-26125905

RESUMEN

We examined whether erythropoietin (EPO) can inhibit adipogenic differentiation of mesenchymal stem cells (MSCs) in the mouse bone marrow and its underlying mechanism. We separated and extracted mouse bone marrow MSCs and induced adipogenic differen-tiation using 3-isobutyl-1-methylxanthine, insulin, and dexamethasone. Different concentrations of EPO were added to the cells and observed by Oil Red O staining on the 20th day to quantitatively analyze the degree of cell differentiation. mRNA expression levels of peroxysome proliferator-activated receptor γ (PPARγ), CCAAT enhancer binding protein α, and adiponectin were analyzed by real-time quantitative polymerase chain reaction, and the activity of PPARγ, extracellular sig-nal-regulated kinase (ERK), and p38 mitogen-activated protein kinase (p38 MAPK) were determined by western blotting. EPO significantly inhibited adipogenic differentiation of MSCs after 20 days and reduced absorbance values by Oil Red O staining without affecting proliferation activity. EPO downregulated the mRNA expression of PPARγ, CCAAT enhancer binding protein α, fatty acid binding protein 4, and adiponec-tin during adipogenesis and increased protein phosphorylation of ERK, p38 MAPK, and PPARγ during differentiation. EPO downregulated the mRNA expression of PPARγ, CCAAT enhancer binding protein α, fatty acid binding protein 4, and adiponectin by increasing protein phosphor-ylation of ERK, p38 MAPK, and PPARγ during differentiation, which inhibited adipogenic differentiation of MSCs.


Asunto(s)
Adipocitos/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Eritropoyetina/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , 1-Metil-3-Isobutilxantina/antagonistas & inhibidores , 1-Metil-3-Isobutilxantina/farmacología , Adipocitos/citología , Adipocitos/enzimología , Adiponectina/genética , Adiponectina/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/enzimología , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular , Dexametasona/antagonistas & inhibidores , Dexametasona/farmacología , Activación Enzimática/efectos de los fármacos , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Insulina/farmacología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/enzimología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/genética
16.
Eur Rev Med Pharmacol Sci ; 19(7): 1130-40, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25912570

RESUMEN

OBJECTIVE: Intrauterine glucocorticoid (GC) exposure is associated with disturbances in feto-placental growth. This study aimed to investigate whether folic acid supplementation can prevent dexamethasone (Dex)-induced feto-placental growth restriction. MATERIALS AND METHODS: Female C57BL/6J mice were subject to four different treatments, respectively: normal drinking water plus saline injection (NN), normal drinking water plus Dex injection (ND), drinking water supplemented with folic acid plus saline injection (FN), and drinking water supplemented with folic acid plus Dex injection (FD). Folic acid (100 µg/L) was administrated since 2 weeks before the mating and throughout pregnancy. Dex injection (100 µg/kg•d) was performed from E12.5 to E16.5. The placentas were collected at E17.5. RESULTS: The parameters including placental and fetal weight, the maximum placental diameter, volume of junctional and labyrinthine zones, and vascular density in the ND group were significantly smaller compared to the NN group. Except the maximum placental diameter, each of the above parameters in the FD group was significantly larger compared to the ND group. The levels of glucocorticoid receptor (GR) protein, and endothelial growth factor A, C (VEGFA, VEGFC) and placental growth factor (PIGF) mRNAs were significantly lower in the ND group compared to NN group. The VEGFA and PIGF mRNA level in the FD group was significantly higher than that in the ND group, as well as VEGFA and VEGFC protein level. CONCLUSIONS: Folic acid may attenuate Dex-induced restriction on placental growth by elevating the expression of VEGFA and PIGF, and further raising vascular density.


Asunto(s)
Dexametasona/toxicidad , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/prevención & control , Ácido Fólico/uso terapéutico , Placenta/efectos de los fármacos , Placentación , Animales , Dexametasona/antagonistas & inhibidores , Suplementos Dietéticos , Femenino , Retardo del Crecimiento Fetal/metabolismo , Ácido Fólico/farmacología , Ratones , Ratones Endogámicos C57BL , Placenta/metabolismo , Embarazo , ARN Mensajero/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis
17.
Biochem Biophys Res Commun ; 454(1): 42-7, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25445596

RESUMEN

Long-time glucocorticoids (GCs) usage causes osteoporosis. In the present study, we explored the potential role of hydrogen sulfide (H2S) against dexamethasone (Dex)-induced osteoblast cell damage, and focused on the underlying mechanisms. We showed that two H2S-producing enzymes, cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CSE), were significantly downregulated in human osteonecrosis tissues as well as in Dex-treated osteoblastic MC3T3-E1 cells. H2S donor NaHS as well as the CBS activator S-adenosyl-l-methionine (SAM) inhibited Dex-induced viability reduction, death and apoptosis in MC3T3-E1 cells. NaHS activated adenosine monophosphate (AMP)-activated protein kinase (AMPK) signaling, which participated its cyto-protective activity. AMPK inhibition by its inhibitor (compound C) or reduction by targeted-shRNA suppressed its pro-survival activity against Dex in MC3T3-E1 cells. Further, we found that NaHS inhibited Dex-mediated reactive oxygen species (ROS) production and ATP depletion. Such effects by NaHS were again inhibited by compound C and AMPKα1-shRNA. In summary, we show that H2S inhibits Dex-induced osteoblast damage through activation of AMPK signaling. H2S signaling might be further investigated as a novel target for anti-osteoporosis treatment.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Dexametasona/antagonistas & inhibidores , Dexametasona/toxicidad , Sulfuro de Hidrógeno/farmacología , Osteoblastos/efectos de los fármacos , Células 3T3 , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Citoprotección/efectos de los fármacos , Activación Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Osteoblastos/metabolismo , Osteoblastos/patología , Osteonecrosis/metabolismo , Osteonecrosis/patología , Osteonecrosis/prevención & control , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo
18.
Br J Pharmacol ; 171(3): 735-45, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24172143

RESUMEN

BACKGROUND AND PURPOSE: The COX-2/PGE2 pathway in hypoxic cancer cells has important implications for stimulation of inflammation and tumourigenesis. However, the mechanism by which glucocorticoid receptors (GRs) inhibit COX-2 during hypoxia has not been elucidated. Hence, we explored the mechanisms underlying glucocorticoid-mediated inhibition of hypoxia-induced COX-2 in human distal lung epithelial A549 cells. EXPERIMENTAL APPROACH: The expressions of COX-2 and glucocorticoid-induced leucine zipper (GILZ) in A549 cells were determined by Western blot and/or quantitative real time-PCR respectively. The anti-invasive effect of GILZ on A549 cells was evaluated using the matrigel invasion assay. KEY RESULTS: The hypoxia-induced increase in COX-2 protein and mRNA levels and promoter activity were suppressed by dexamethasone, and this effect of dexamethasone was antagonized by the GR antagonist RU486. Overexpression of GILZ in A549 cells also inhibited hypoxia-induced COX-2 expression levels and knockdown of GILZ reduced the glucocorticoid-mediated inhibition of hypoxia-induced COX-2 expression, indicating that the inhibitory effects of dexamethasone on hypoxia-induced COX-2 are mediated by GILZ. GILZ suppressed the expression of hypoxia inducible factor (HIF)-1α at the protein level and affected its signalling pathway. Hypoxia-induced cell invasion was also dramatically reduced by GILZ expression. CONCLUSION AND IMPLICATIONS: Dexamethasone-induced upregulation of GILZ not only inhibits the hypoxic-evoked induction of COX-2 expression and cell invasion but further blocks the HIF-1 pathway by destabilizing HIF-1α expression. Taken together, these findings suggest that the suppression of hypoxia-induced COX-2 by glucocorticoids is mediated by GILZ. Hence, GILZ is a potential key therapeutic target for suppression of inflammation under hypoxia.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Receptores de Glucocorticoides/agonistas , Factores de Transcripción/agonistas , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/metabolismo , Animales , Antiinflamatorios no Esteroideos/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/farmacología , Hipoxia de la Célula , Línea Celular , Movimiento Celular/efectos de los fármacos , Ciclooxigenasa 2/química , Ciclooxigenasa 2/genética , Dexametasona/antagonistas & inhibidores , Dexametasona/farmacología , Genes Reporteros/efectos de los fármacos , Glucocorticoides/antagonistas & inhibidores , Antagonistas de Hormonas/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Interferencia de ARN , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Neuropsychopharmacology ; 38(10): 2068-79, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23648781

RESUMEN

Prenatal stress or exposure to elevated levels of glucocorticoids (GCs) can impair specific neurobehavioral circuits leading to alterations in emotional processes later in life. In turn, emotional deficits may interfere with the quality and degree of social interaction. Here, by using a comprehensive behavioral approach in combination with the measurement of ultrasonic vocalizations, we show that in utero GC (iuGC)-exposed animals present increased immobility in the forced swimming test, pronounced anhedonic behavior (both anticipatory and consummatory), and an impairment in social interaction at different life stages. Importantly, we also found that social behavioral expression is highly dependent on the affective status of the partner. A profound reduction in mesolimbic dopaminergic transmission was found in iuGC animals, suggesting a key role for dopamine (DA) in the etiology of the observed behavioral deficits. Confirming this idea, we present evidence that a simple pharmacological approach-acute L-3,4-dihydroxyphenylacetic acid (L-DOPA) oral administration, is able to normalize DA levels in iuGC animals, with a concomitant amelioration of several dimensions of the emotional and social behaviors. Interestingly, L-DOPA effects in control individuals were not so straightforward; suggesting that both hypo- and hyperdopaminergia are detrimental in the context of such complex behaviors.


Asunto(s)
Afecto/fisiología , Dexametasona/efectos adversos , Neuronas Dopaminérgicas/fisiología , Glucocorticoides/efectos adversos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/psicología , Conducta Social , Afecto/efectos de los fármacos , Factores de Edad , Anhedonia/efectos de los fármacos , Anhedonia/fisiología , Animales , Dexametasona/administración & dosificación , Dexametasona/antagonistas & inhibidores , Neuronas Dopaminérgicas/efectos de los fármacos , Femenino , Glucocorticoides/administración & dosificación , Glucocorticoides/antagonistas & inhibidores , Pérdida de Tono Postural/efectos de los fármacos , Pérdida de Tono Postural/fisiología , Levodopa/farmacología , Sistema Límbico/efectos de los fármacos , Sistema Límbico/fisiología , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ratas , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Vocalización Animal/efectos de los fármacos , Vocalización Animal/fisiología
20.
Mol Psychiatry ; 18(3): 300-7, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23380766

RESUMEN

Neurogenesis during embryonic and adult life is tightly regulated by a network of transcriptional, growth and hormonal factors. Emerging evidence indicates that activation of the stress response, via the associated glucocorticoid increase, reduces neurogenesis and contributes to the development of adult diseases.As corticotrophin-releasing hormone (CRH) or factor is the major mediator of adaptive response to stressors, we sought to investigate its involvement in this process. Accordingly, we found that CRH could reverse the damaging effects of glucocorticoid on neural stem/progenitor cells (NS/PCs), while its genetic deficiency results in compromised proliferation and enhanced apoptosis during neurogenesis. Analyses in fetal and adult mouse brain revealed significant expression of CRH receptors in proliferating neuronal progenitors. Furthermore, by using primary cultures of NS/PCs, we characterized the molecular mechanisms and identified CRH receptor-1 as the receptor mediating the neuroprotective effects of CRH. Finally, we demonstrate the expression of CRH receptors in human fetal brain from early gestational age, in areas of active neuronal proliferation. These observations raise the intriguing possibility for CRH-mediated pharmacological applications in diseases characterized by altered neuronal homeostasis, including depression, dementia, neurodegenerative diseases, brain traumas and obesity.


Asunto(s)
Encéfalo/efectos de los fármacos , Hormona Liberadora de Corticotropina/farmacología , Neurogénesis/fisiología , Fármacos Neuroprotectores/farmacología , Células Madre/fisiología , Animales , Apoptosis/fisiología , Encéfalo/metabolismo , Proliferación Celular/efectos de los fármacos , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Dexametasona/antagonistas & inhibidores , Dexametasona/toxicidad , Humanos , Ratones , Ratones Noqueados , Neurogénesis/efectos de los fármacos , Pirimidinas/farmacología , Pirroles/farmacología , Receptores de Hormona Liberadora de Corticotropina/agonistas , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Hormona Liberadora de Corticotropina/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA