Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 942
Filtrar
1.
Behav Brain Res ; 418: 113664, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-34780858

RESUMEN

Recreational abuse of solvents continues, despite cyclohexane (CHX) is used as a safe replacement in gasoline or adhesive formulations. Increasing evidence indicates that CHX inhalation affects brain functioning; however, scanty information is available about its effects on behavior and brain activity upon drug removal. In this study, we used CD1 adult mice to mimic an intoxication period of recreational drugs for 30 days. During the CHX exposure (~30,000 ppm), we analyzed exploratory and biphasic behaviors, stereotypic circling, and locomotion. After CHX removal (24 h or a month later), we assessed anxiety-like behaviors and quantified c-Fos cells in motor- and anxiety-related brain regions. Our findings indicate that the repeated inhalation of CHX produced steady hyperactivity and reduced ataxia, sedation, and seizures as the exposure to CHX progressed. Also, CHX decreased grooming and rearing behaviors. In the first week of CHX inhalation, a stereotypic circling behavior emerged, and locomotion increased gradually. One month after CHX withdrawal, mice showed low activity in the center zone of the open field and more buried marbles. Twenty-four hours after CHX removal, c-Fos expression was low in the dorsal striatum, ventral striatum, motor cortex, dorsomedial prefrontal cortex, basolateral amygdala, lateral hypothalamus, and ventral hippocampus. One month later, c-Fos expression remained low in the ventral striatum and lateral hypothalamus but increased in the dorsomedial prefrontal cortex and primary motor cortex. This study provides a comprehensive behavioral characterization and novel histological evidence of the CHX effects on the brain when is administered in a recreational-like mode.


Asunto(s)
Ansiedad/fisiopatología , Ciclohexanos , Conducta Exploratoria/efectos de los fármacos , Hipercinesia/fisiopatología , Exposición por Inhalación/efectos adversos , Locomoción/efectos de los fármacos , Animales , Ciclohexanos/metabolismo , Ciclohexanos/farmacología , Genes fos/genética , Masculino , Ratones , Corteza Motora/metabolismo , Corteza Prefrontal/metabolismo , Estriado Ventral/metabolismo
2.
Clin Exp Pharmacol Physiol ; 49(3): 341-349, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34729812

RESUMEN

An imbalance of osteoclasts and osteoblasts can result in a variety of bone-related diseases, including osteoporosis. Thus, decreasing the activity of osteoclastic bone resorption is the main therapeutic method for treating osteoporosis. 2E-Decene-4, 6-diyn-1-ol-acetate (DDA) is a natural bioactive compound with anti-inflammatory and anti-cancer properties. However, its effects on osteoclastogenesis are unknown. Murine bone marrow-derived macrophages (BMMs) or RAW264.7 cells were treated with DDA, followed by evaluation of cell viability, RANKL-induced osteoclast differentiation, and pit formation assay. Effects of DDA on RANKL-induced phosphorylation of MAPKs were assayed by western blot analysis. Expression of osteoclast-specific genes was examined with reverse transcription-PCR (RT-PCR) and western blot analysis. In this study, DDA significantly inhibited RANKL-induced osteoclast differentiation in RAW264.7 cells as well as in BMMs without cytotoxicity. DDA also strongly blocked the resorbing capacity of BMM on calcium phosphate-coated plates. DDA inhibited RANKL-induced phosphorylation of ERK, JNK and p38 MAPKs, as well as expression of c-Fos and NFATc1, which are essential transcription factors for osteoclastogenesis. In addition, DDA decreased expression levels of osteoclastogenesis-specific genes, including matrix metalloproteinase-9 (MMP-9), tartrate-resistant acid phosphatase (TRAP), and receptor activator of NF-κB (RANK) in RANKL-induced RAW264.7 cells. Collectively, these findings indicated that DDA attenuates RANKL-induced osteoclast formation by suppressing the MAPKs-c-Fos-NFATc1 signalling pathway and osteoclast-specific genes. These results indicate that DDA may be a potential candidate for bone diseases associated with abnormal osteoclast formation and function.


Asunto(s)
Productos Biológicos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genes fos/fisiología , Macrófagos/efectos de los fármacos , Factores de Transcripción NFATC/metabolismo , Osteogénesis/efectos de los fármacos , Animales , Aster/química , Productos Biológicos/química , Diferenciación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genes fos/genética , Ratones , Factores de Transcripción NFATC/genética , Osteoclastos , Ligando RANK/genética , Ligando RANK/metabolismo , Células RAW 264.7
3.
Aging (Albany NY) ; 13(15): 19878-19893, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34375303

RESUMEN

To date, different experimental strategies have been developed for the ex vivo expansion of human hematopoietic stem cells (HSCs) for clinical applications. However, differences in the genomic function of expanded HSCs under different culture systems remain unclear. In this study, we compared the gene expression profiles of HSCs in ex vivo expanded serum (10% FBS, fetal bovine serum) and serum-free culture systems and analyzed the molecular functions of differentially expressed genes using microarray chips. We identified 839 differentially expressed genes between the two culture systems. These genes were enriched in the TNF -regulated inflammatory pathway in an FBS culture system. In addition, the mRNA expression of CCL2 (C-C motif chemokine ligand 2), TNF (tumor necrosis factor) and FOS (FBJ murine osteosarcoma viral oncogene homolog) was validated by RT-qPCR. Our data revealed that ex vivo expansion of HSCs using the FBS culture system induces an inflammatory response and high CD38 expression, indicating that this system might activate an inflammatory pathway and induce expression of the cancer marker CD38 during ex vivo expansion of HSCs. This study provides a transcriptional profile and new insights into the genomic functions of HSCs under different expanded cultures.


Asunto(s)
Antígenos CD34/metabolismo , Quimiocina CCL2/metabolismo , Sangre Fetal/citología , Células Madre Hematopoyéticas/fisiología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Cultivadas , Quimiocina CCL2/genética , Perfilación de la Expresión Génica , Genes fos/genética , Humanos , Recién Nacido , Factor de Necrosis Tumoral alfa/genética
4.
Environ Toxicol Pharmacol ; 84: 103607, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33545377

RESUMEN

Human bronchial epithelial (HBE) cells and c-fos-silenced HBE cells were first exposed to fine particulate matter (PM2.5) and the resulting miRNA sequenced. Thereafter, a weighted gene co-expression network analysis was performed using Cytoscape software to visualize the interactions between identified hub miRNAs and their target genes. Nine differentially expressed miRNAs in hub miRNAs were identified in the different treatment groups, of which miR-25-3p, miR-215-5p, and miR-145-5p were selected for further study. Following qPCR validation, both miR-25-3p and miR-215-5p were found to be significantly up-regulated whilst, miR-145-5p was significantly down-regulated (p < 0.05) in the PM2.5 group. Furthermore, miR-25-3p and miR-145-5p were also significantly down-regulated in the untreated group of c-fos silenced HBE cells. However, miR-215-5p was significantly down-regulated in both the untreated and PM2.5-treated groups of c-fos silenced HBE cells. Subsequent analysis of their target genes also illustrated differential gene expression when comparing the treatment groups of the two cell types. The present data indicated that the c-fos gene has an important effect on the miRNA expression profiles and the related signaling pathways in PM2.5-treated HBE cells. Therefore, each of miR-25-3p, miR-145-5p, and miR-215-5p may potentially provide future research information for additional exploration of a PM2.5-induced carcinogenesis mechanism.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Células Epiteliales/efectos de los fármacos , Genes fos/genética , MicroARNs , Material Particulado/toxicidad , Bronquios/citología , Línea Celular , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos
5.
Int J Mol Med ; 47(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33604677

RESUMEN

The Fos proto­oncogene, activator protein­1 (AP­1) transcription factor subunit (c­fos) gene, a member of the immediate early gene family, encodes c­Fos, which is a subunit of the AP­1 transcription factor. The present study aimed to investigate the mechanism by which the translation efficiency of c­fos mRNA is upregulated when cellular protein synthesis is shut off. The result of western blotting revealed that the protein expression levels of c­Fos were increased in rhabdomyosarcoma cells infected with enterovirus 71 (EV71) compared with uninfected cells. PCR was used to get the c­fos 5'­untranslated region (UTR). The luciferase assay of a bicistronic vector containing the c­fos 5'UTR revealed that the c­fos 5'UTR contains an internal ribosome entry site (IRES) sequence and a 175 nucleotide sequence (between 31 and 205 nt) that is essential for IRES activity. Analysis of potential IRES trans­acting factors revealed that poly(C)­binding protein 2 (PCBP2) negatively regulated the activity of the c­fos IRES, whereas the La autoantigen (La) positively regulated its activity. The results of RNA­protein immunoprecipitation demonstrated that both PCBP2 and La bound to the c­fos 5'UTR. Furthermore, the IRES activity of in vitro­transcribed c­fos mRNA was upregulated during EV71 infection. The present study suggested a mechanism for the effect of viral infection on host genes, and provided a novel target for gene translation regulation.


Asunto(s)
Regiones no Traducidas 5'/genética , Regulación de la Expresión Génica/genética , Genes fos/genética , Sitios Internos de Entrada al Ribosoma/genética , Proteínas Proto-Oncogénicas c-fos/genética , Autoantígenos/metabolismo , Secuencia de Bases/genética , Línea Celular Tumoral , Enterovirus Humano A/metabolismo , Células HEK293 , Células HeLa , Humanos , Biosíntesis de Proteínas/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/virología , Ribonucleoproteínas/metabolismo , Ribosomas/metabolismo , Factor de Transcripción AP-1/genética , Transcripción Genética/genética , Antígeno SS-B
6.
J Neurochem ; 157(3): 532-549, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33454999

RESUMEN

Interleukin-6 (IL-6) is a cytokine primarily known for immune regulation. There is also growing evidence that IL-6 triggers neurogenesis and impacts neural development, both life-long occurring processes that can be impaired by early-life and adult stress. Stress induces the release of glucocorticoids by activation of the hypothalamic-pituitary-adrenal (HPA) axis. On the cellular level, glucocorticoids act via the ubiquitously expressed glucocorticoid receptor. Thus, we aimed to elucidate whether glucocorticoids affect IL-6-induced neural development. Here, we show that IL-6 signalling induces neurite outgrowth in adrenal pheochromocytoma PC12 cells in a mitogen-activated protein kinase (MAPK) pathway-dependent manner, since neurite outgrowth was diminished upon Mek-inhibitor treatment. Using quantitative biochemical approaches, such as qRT-PCR analysis of Hyper-IL-6 treated PC12 cells, we show that neurite outgrowth induced by IL-6 signalling is accompanied by early and transient MAPK-dependent mRNA expression of immediate early genes coding for proteins such as early growth response protein 1 (Egr1) and c-Fos. This correlates with reduced proliferation and prolonged G0/G1 cell cycle arrest as determined by monitoring the cellular DNA content using flow cytometry. These results indicate for IL-6 signalling-induced neural differentiation. Interestingly, the glucocorticoid Dexamethasone impairs early IL-6 signalling-induced mRNA expression of c-Fos and Egr1 and restrains neurite outgrowth. Impaired Egr1 and c-Fos expression in neural development is implicated in the aetiology of neuropathologies. Thus, it appears likely that stress-induced release of glucocorticoids, as well as therapeutically administered glucocorticoids, contribute to the development of neuropathologies by reducing the expression of Egr1 and c-Fos, and by restraining IL-6-dependent neural differentiation.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Genes fos/efectos de los fármacos , Genes fos/genética , Glucocorticoides/farmacología , Interleucina-6/antagonistas & inhibidores , Neuritas/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dexametasona/farmacología , Glucocorticoides/antagonistas & inhibidores , Antagonistas de Hormonas/farmacología , Mifepristona/farmacología , Células PC12 , Ratas
7.
Endocrinology ; 162(1)2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33034617

RESUMEN

AbstractCentral 5-hydroxytryptamine (5-HT), which is primarily synthesized by tryptophan hydroxylase 2 (TPH2) in the dorsal Raphe nuclei (DRN), plays a pivotal role in the regulation of food intake and body weight. However, the physiological functions of TPH2 on energy balance have not been consistently demonstrated. Here we systematically investigated the effects of TPH2 on energy homeostasis in adult male and female mice. We found that the DRN harbors a similar amount of TPH2+ cells in control male and female mice. Adult-onset TPH2 deletion in the DRN promotes hyperphagia and body weight gain only in male mice, but not in female mice. Ablation of TPH2 reduces hypothalamic pro-opiomelanocortin (POMC) neuronal activity robustly in males, but only to a modest degree in females. Deprivation of estrogen by ovariectomy (OVX) causes comparable food intake and weight gain in female control and DRN-specific TPH2 knockout mice. Nevertheless, disruption of TPH2 blunts the anorexigenic effects of exogenous estradiol (E2) and abolishes E2-induced activation of POMC neurons in OVX female mice, indicating that TPH2 is indispensable for E2 to activate POMC neurons and to suppress appetite. Together, our study revealed that TPH2 in the DRN contributes to energy balance regulation in a sexually dimorphic manner.


Asunto(s)
Núcleo Dorsal del Rafe/metabolismo , Metabolismo Energético/fisiología , Triptófano Hidroxilasa/metabolismo , Aumento de Peso/genética , Animales , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Genes fos/genética , Genes fos/fisiología , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Ovariectomía , Factores Sexuales , Triptófano Hidroxilasa/genética , Aumento de Peso/fisiología
8.
J Psychopharmacol ; 35(4): 483-493, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33143539

RESUMEN

BACKGROUND: Psilocybin is a serotonergic psychedelic found in "magic mushrooms" with a putative therapeutic potential for treatment-resistant depression, anxiety, obsessive-compulsive disorder, and addiction. In rodents, psilocybin acutely induces plasticity-related immediate early genes in cortical tissue; however, studies into the effects on subcortical regions, of different doses, and the subsequent translation of corresponding proteins are lacking. METHODS: We examined the acute effects of a single administration of psilocybin (0.5-20 mg/kg) on the expression of selected genes in the prefrontal cortex and hippocampus. In total, 46 target genes and eight reference genes were assessed using real-time quantitative polymerase chain reaction. Corresponding protein levels of the three most commonly regulated genes were assessed using Western blotting. RESULTS: In the prefrontal cortex, psilocybin increased the expression of Cebpb, c-Fos, Dups1, Fosb, Junb, Iκß-α, Nr4a1, P11, Psd95, and Sgk1, and decreased the expression of Clk1. In the hippocampus, psilocybin strongly increased the expression of Arrdc2, Dusp1, Iκß-α, and Sgk1 in a dose-dependent manner, and decreased the expression of Arc, Clk1, Egr2, and Ptgs2. Protein levels of Sgk1, Dusp1, and Iκß-α showed only partial agreement with transcriptional patterns, stressing the importance of assessing downstream translation when investigating rapid gene responses. CONCLUSION: The present study demonstrates that psilocybin rapidly induces gene expression related to neuroplasticity, biased towards the prefrontal cortex, compared to the hippocampus. Our findings provide further evidence for the rapid plasticity-promoting effects of psilocybin.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo , Plasticidad Neuronal , Corteza Prefrontal , Psilocibina/farmacología , Animales , Proteínas del Citoesqueleto/genética , Relación Dosis-Respuesta a Droga , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Genes Inmediatos-Precoces , Genes fos/genética , Alucinógenos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Biomed Environ Sci ; 33(9): 680-689, 2020 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-33106213

RESUMEN

OBJECTIVE: To investigate the effect of c-fos gene silencing on differentially expressed proteins (DEPs) in human bronchial epithelial (HBE) cells after exposure to fine particulate matter (PM 2.5). METHODS: HBE cells and c-fos-silenced HBE cells were exposed to 50 µg/mL PM 2.5, LC-MS/MS and tandem mass tag (TMT) labeling methods were combined with bioinformatics methods, and DEPs and interaction networks were identified. RESULTS: In the HBE group, 414 DEPs were screened, of which 227 were up-regulated and 187 down-regulated. In the c-fos silenced HBE group, 480 DEPs were screened, including 240 up-regulated proteins and 240 down-regulated proteins. KEGG annotations showed that DEPs in the HBE group are mainly concentrated in the glycolysis/gluconeogenesis pathway and those in the c-fos silenced group are concentrated mainly in endoplasmic reticulum and the processing of proteins. Additionally, the abnormal expression of GPRC5C, DKK4, and UBE2C was identified in top 15 DEPs. After constructing the protein interaction network, 20 Hub proteins including HNRNPA2B1, HNRNPL, RPS15A, and RPS25 were screened from the HBE group and the c-fos silenced HBE group. CONCLUSION: c-fos gene affected the expression of cancer-related proteins. Our results provided a scientific basis for further study of PM 2.5-induced carcinogenesis mechanism.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Bronquios/efectos de los fármacos , Regulación de la Expresión Génica , Silenciador del Gen , Genes fos/genética , Material Particulado/toxicidad , Mucosa Respiratoria/efectos de los fármacos , Bronquios/metabolismo , Células Cultivadas , Humanos , Proteómica , Mucosa Respiratoria/metabolismo
10.
Cell Mol Neurobiol ; 40(8): 1395-1404, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32162199

RESUMEN

In the present work, using in situ hybridization, we studied the expression patterns of three molluscan homologs of vertebrate immediate-early genes C/EBP, c-Fos, and c-Jun in the central nervous system (CNS) of terrestrial gastropod snail Helix. The molluscan C/EBP gene was described in literature, while c-Fos and c-Jun were studied in terrestrial snails for the first time. Localization of the expression was traced in normal conditions, and in preparations physiologically activated using stimulation of suboesophageal ganglia nerves. No expression was detected constitutively. In stimulated preparations, all three genes had individual expression patterns in Helix CNS, and the level of expression was stimulus-dependent. The number of cells expressing the gene of interest was different from the number of cells projecting to the stimulated nerve, and thus activated retrogradely. This difference depended on the ganglia studied. At the subcellular level, the labeled RNA was observed as dots (probably small clusters of RNA molecules) and shapeless mass of RNA, often seen as a circle at the internal border of the cell nuclei. The data provide a basis for further study of behavioral role of these putative immediate-early genes in snail behavior and learning.


Asunto(s)
Sistema Nervioso Central/metabolismo , Genes Inmediatos-Precoces/genética , Neuronas/metabolismo , Caracoles/genética , Animales , Genes Inmediatos-Precoces/fisiología , Genes fos/genética , Caracoles Helix/fisiología , Proteínas Proto-Oncogénicas c-fos/biosíntesis , ARN/metabolismo
11.
J Steroid Biochem Mol Biol ; 199: 105594, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31968225

RESUMEN

Breast cancer is currently the leading cause of cancer death among women worldwide. AP-1 (c-Fos/c-Jun) is associated with proliferation and survival, while cytoplasmic c-Fos activates phospholipid synthesis in cells induced to differentiate or grow. Estrogen receptor α 46 (ERα46) is a splice variant of full-length ERα66 and it is known that it has an inhibitory role in cancer cell growth. We investigated c-Fos localization, its relationship to AP-1, the non genomic pathway of phospho-Tyr537-ERα66, as well as ERα46 and ERα66 isoforms in rat mammary gland development and carcinogenic transformation, and in mammary tumors. Female rats were injected: a) saline solution (Control mammary gland, CMG) or b) N-Nitroso-N-methyl urea (NMU), and samples were taken at 60, 90, 120 and 150 days of life. In addition, we analyzed hormone-dependent (HD) and independent (HI) tumors in ovariectomized rats, and intact tumors (IT) in non-ovariectomized ones. Our results show that, in CMG, nuclear c-Fos and proliferation decreased with age, AP-1 content was low, and nuclear ERα46/ERα66 ratio was higher than 1. In NMU, nuclear c-Fos and proliferation increased with carcinogenic transformation, AP-1 content was high, and nuclear ERα46/ERα66 was below 1. As tumor grade increased, proliferation, nuclear c-Fos and AP-1 expression were negatively associated to nuclear ERα46/ERα66 in IT. In HD, nuclear ERα46/ERα66, nuclear c-Fos expression, AP-1 levels and proliferation were lower than in HI, whose growth is estrogen-independent. Phospho-Tyr537-ERα66 content and ERK1/2 activation were associated with AP-1 levels and cell proliferation. Collectively, our findings support the notion that variant detection and ERα46/ERα66 ratio could shed light on the role of ERα isoforms in mammary gland transformation and the behavior of ERα positive mammary tumors.


Asunto(s)
Receptor alfa de Estrógeno/genética , Genes fos/genética , Neoplasias Mamarias Animales/genética , Isoformas de Proteínas/genética , Animales , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Proliferación Celular/efectos de los fármacos , Citoplasma/efectos de los fármacos , Citoplasma/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Metilnitrosourea/farmacología , Isoformas de Proteínas/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/genética
12.
Neuropharmacology ; 164: 107912, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31843397

RESUMEN

Post-traumatic stress disorder (PTSD) is a severe chronic mental illness that develops in individuals exposed to life-threatening trauma and is characterized by hyperarousal, flashbacks and nightmares. The serotonergic (5-HT) and noradrenergic (NE) systems are deeply involved in the pathogenesis of PTSD. We have previously reported a novel anxiolytic compound, ACH-000029, that modulates 5-HT and α1-adrenergic receptors and induces acute anxiolytic-like effects in rodents. Here, we investigated the potential of ACH-000029 to prevent anxiety-like behavior in the single prolonged stress (SPS) PTSD model. Mice were subjected to the SPS procedure, followed by a 7-day treatment with ACH-000029 and, for comparison, with the α1-adrenergic antagonist prazosin. Animals were behaviorally assessed using social interaction, elevated plus maze and open field tests. Interestingly, treatment with ACH-000029 but not with prazosin ameliorated the SPS-induced sociability impairment and anxiety-like behavior. The brain-wide c-fos mapping, used as a surrogate for brain activity, indicated the brain structures that were altered by SPS and putatively involved in the anxiolytic-like effect of ACH-000029. The SPS protocol produced long-lasting impairment of regions involved in stress-anxiety response, such as the amygdala, prefrontal cortex, globus pallidus and superior colliculus. ACH-000029 treatment reversed the SPS-induced c-fos changes in the globus pallidus, lateral septum and entorhinal cortex and exclusively modulated c-fos levels in subregions from the retrosplenial cortex, cerebellum, superior colliculus and ventromedial hypothalamus. These results support the hypothesis that the dual regulation of 5-HT and α1-adrenergic receptors is required to alleviate PTSD symptoms and suggest a possible role of ACH-000029 as a PTSD treatment.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Piperazinas/farmacología , Quinazolinas/farmacología , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Serotoninérgicos/farmacología , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/psicología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Ansiolíticos/uso terapéutico , Química Encefálica/efectos de los fármacos , Genes fos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Piperazinas/uso terapéutico , Prazosina/farmacología , Quinazolinas/uso terapéutico , Serotoninérgicos/uso terapéutico , Interacción Social , Estrés Psicológico/complicaciones , Estrés Psicológico/psicología
13.
Genes Dev ; 34(1-2): 72-86, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31831627

RESUMEN

Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ) are key effectors of the Hippo pathway to control cell growth and organ size, of which dysregulation yields to tumorigenesis or hypertrophy. Upon activation, YAP/TAZ translocate into the nucleus and bind to TEAD transcription factors to promote transcriptional programs for proliferation or cell specification. Immediate early genes, represented by AP-1 complex, are rapidly induced and control later-phase transcriptional program to play key roles in tumorigenesis and organ maintenance. Here, we report that YAP/TAZ directly promote FOS transcription that in turn contributes to the biological function of YAP/TAZ. YAP/TAZ bind to the promoter region of FOS to stimulate its transcription. Deletion of YAP/TAZ blocks the induction of immediate early genes in response to mitogenic stimuli. FOS induction contributes to expression of YAP/TAZ downstream target genes. Genetic deletion or chemical inhibition of AP-1 suppresses growth of YAP-driven cancer cells, such as Lats1/2-deficient cancer cells as well as Gαq/11 mutated uveal melanoma. Furthermore, AP-1 inhibition almost completely abrogates the hepatomegaly induced by YAP overexpression. Our findings reveal a feed-forward interplay between immediate early transcription of AP-1 and Hippo pathway function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación Neoplásica de la Expresión Génica , Transactivadores/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes fos/genética , Células HEK293 , Humanos , Hígado/metabolismo , Melanoma/fisiopatología , Ratones , Mitógenos/farmacología , Tamaño de los Órganos/genética , Regiones Promotoras Genéticas/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Neoplasias de la Úvea/fisiopatología , Proteínas Señalizadoras YAP
14.
Brain Res Bull ; 153: 102-108, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31445055

RESUMEN

Neural circuits involved in the development of depression are currently poorly understood. To provide insight into this issue, we evaluated the influence of seven clinically effective antidepressants on neuronal activity in thirty rat brain areas. Drugs belonging to all major groups of antidepressants (imipramine, reboxetine, fluoxetine, bupropion, mirtazapine, agomelatine, and phenelzine) were examined; since antidepressants typically require weeks of continued administration before they achieve a therapeutic effect, we administered these drugs for 21 days. The experiments were conducted with male Wistar rats. To identify the neuroanatomical targets for antidepressants, the alterations of c-Fos expression in different brain areas were measured using ELISA assay. The drugs were examined at doses sufficient to produce behavioral effect in the rat forced swim test (FST). All the drugs at the behaviorally relevant doses activated two brain areas, the lateral entorhinal cortex and dorsal subiculum of the hippocampus; none of the drugs affected the c-Fos expression in the medial orbital, prelimbic and infralimbic cortex, caudate putamen, nucleus accumbens core, bed nucleus of stria terminalis, hipothalamic paraventricular nucleus, medial amygdaloid nucleus, lateral habenula, substantia nigra pars compacta and pars reticulata, ventral tegmental area, hippocampal ventral subiculum, dorsal and ventral periaqueductal gray matters, and medial entorhinal cortex. These findings suggest that the stimulation of the lateral entorhinal cortex and hippocampal dorsal subiculum play a role in therapeutic effects of antidepressants.


Asunto(s)
Antidepresivos/metabolismo , Antidepresivos/farmacología , Genes fos/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Depresión/tratamiento farmacológico , Depresión/metabolismo , Corteza Entorrinal/efectos de los fármacos , Corteza Entorrinal/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Genes fos/genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar
15.
Neurochem Int ; 131: 104521, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31419453

RESUMEN

Chronic pain is a worldwide major health problem and many pain-suffering patients are under opioid based therapy. Epidemiological data show that pain intensity correlates with the risk of misuse of prescription opioids, and other drugs of abuse including alcohol. This increased vulnerability to suffer Substance Use Disorders could be, in part, caused by functional changes that occur over the mesocorticolimbic system, a brain pathway involved in reward processing and addiction. Previous data in rats revealed that inflammatory pain desensitizes mu opioid receptors (MORs) in the ventral tegmental area (VTA). As a consequence, pain alters dopamine release in the nucleus accumbens (NAc) derived from MOR activation in the VTA and also increases intake of high doses of heroine. Given that the VTA neurons target different brain regions, in the present study we first analyzed changes induced by inflammatory pain in the MOR dependent activation pattern of the main VTA projecting areas. To do that, we administered two doses (7 or 14 ng) of DAMGO (MORs agonist) or artificial cerebrospinal fluid (aCSF) focally into the VTA of rats and measured the activation in projection areas by cFos immunohistochemistry. Our results show that focal injections of DAMGO in the VTA increases cFos expression in the majority of its projecting areas, namely NAc, basolateral amygdala (BLA), cingulate cortex (ACC) and bed nucleus of the stria terminalis (BNST), as compared to aCSF. Second, we analyzed whether inflammatory pain would affect to cFos expression using a group of rats injected with CFA in the hind paw. In this case, we found that cFos expression was not significantly different between DAMGO and aCSF administered rats in BLA, ACC and BNST. Our results confirm that inflammatory pain induces desensitization of VTA MORs in a region dependent manner which can be very relevant for addictive behaviours.


Asunto(s)
Genes fos/genética , Inflamación/metabolismo , Dolor/metabolismo , Receptores Opioides mu/agonistas , Área Tegmental Ventral/metabolismo , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Adyuvante de Freund , Regulación de la Expresión Génica , Inmunohistoquímica , Inflamación/inducido químicamente , Masculino , Microinyecciones , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Ratas , Área Tegmental Ventral/efectos de los fármacos
16.
Mol Biol Rep ; 46(5): 5425-5432, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31364017

RESUMEN

Obesity is a complex disorder that is influenced by genetic and environmental factors. DNA methylation is an epigenetic mechanism that is involved in development of obesity and its metabolic complications. The aim of this study was to investigate the association between the RANKL and c-Fos gene methylation on obesity with body mass index (BMI), lipid parameters, homeostasis model assessment of insulin resistance (HOMA-IR), plasma leptin, adiponectin and resistin levels. The study included 68 obese and 46 non-obese subjects. Anthropometric parameters, including body weight, body mass index, waist circumference, and waist-hip ratio, were assessed. Serum glucose, triglycerides (TG), total cholesterol, high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C), plasma leptin, adiponectin and resistin levels were measured. Methylation status of RANKL and c-Fos gen were evaluated by MS-HRM. Statistically significant differences were observed between obese patients and the controls with respect to RANKL and c-Fos gene methylation status (p < 0.001). Also, statistically significant importance was observed RANKL gene methylation and increased level of leptin in obese subjects (p = 0.0081). At the same time, statistically significant association between methylation of c-Fos and increased level of adiponectin was observed in obese patients (p = 0.03) On the other hand, decreased level of resistin was observed where the c-Fos was unmetyladed in controls (p = 0.01). We conclude that methylation of RANKL and c-Fos genes have significant influences on obesity and adipokine levels. Based on literature this was the first study which shows the interactions between RANKL and c-Fos methylation and obesity.


Asunto(s)
Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Obesidad/genética , Adiponectina/análisis , Adiponectina/sangre , Adulto , Antropometría , Índice de Masa Corporal , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Metilación de ADN/genética , Femenino , Genes fos/genética , Humanos , Resistencia a la Insulina/genética , Leptina/análisis , Leptina/sangre , Masculino , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/genética , Triglicéridos/sangre
17.
Brain Res Bull ; 152: 74-84, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31279580

RESUMEN

Seizure activity stimulates adult neurogenesis, the birth of new neurons, in the hippocampus. Many new neurons that develop in the presence of repeatedly induced seizures acquire abnormal morphological and functional characteristics that can promote network hyperexcitability and hippocampal dysfunction. However, the impact of seizure induced neurogenesis on behaviour remains poorly understood. In this study, we investigated whether adult-born neurons generated immediately before and during chronic seizures were capable of integration into behaviorally relevant hippocampal networks. Adult rats underwent pentylenetetrazole (PTZ) kindling for either 1 or 2 weeks. Proliferating cells were labelled with BrdU immediately before kindling commenced. Twenty-four hours after receiving their last kindling treatment, rats were placed in a novel environment and allowed to freely explore for 30 min. The rats were euthanized 90 min later to examine for behaviourally-induced immediate early gene expression (c-fos, Zif268). Using this approach, we found that PTZ kindled rats did not differ from control rats in regards to exploratory behaviour, but there was a marked attenuation in behaviour-induced expression of Fos and Zif268 for rats that received 2 weeks of PTZ kindling. Further examination revealed that PTZ kindled rats showed reduced colocalization of Fos and Zif268 in 2.5 week old BrdU + cells. The proportion of immature granule cells (doublecortin-positive) expressing behaviorally induced Zif268 was also significantly lower for PTZ kindled rats than control rats. These results suggest that chronic seizures can potentially disrupt the ability of adult-born cells to functionally integrate into hippocampal circuits important for the processing of spatial information.


Asunto(s)
Conducta Exploratoria/fisiología , Excitación Neurológica/metabolismo , Neurogénesis/fisiología , Pentilenotetrazol/farmacología , Animales , Bromodesoxiuridina/metabolismo , Convulsivantes/farmacología , Proteína Doblecortina , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Conducta Exploratoria/efectos de los fármacos , Genes Inmediatos-Precoces , Genes fos/genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Excitación Neurológica/efectos de los fármacos , Masculino , Neurogénesis/efectos de los fármacos , Neuronas/metabolismo , Pentilenotetrazol/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Transcriptoma/genética
18.
Nutrients ; 11(6)2019 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167465

RESUMEN

Chili peppers are one of the most widely consumed spices worldwide. However, research on the health benefits of chili peppers and some of its constituents has raised controversy as to whether chili pepper compounds possess cancer-promoting or cancer-preventive effects. While ample studies have been carried out to examine the effect of capsaicin in carcinogenesis, the chemopreventive effect of other major components in chili pepper, including dihydrocapsaicin, capsiate, and capsanthin, is relatively unclear. Herein, we investigated the inhibitory effect of chili pepper components on malignant cell transformation. Among the tested chili pepper compounds, dihydrocapsaicin displayed the strongest inhibitory activity against epidermal growth factor (EGF)-induced neoplastic transformation. Dihydrocapsaicin specifically suppressed EGF-induced phosphorylations of the p70S6K1-S6 pathway and the expression of c-Fos. A reduction in c-Fos levels by dihydrocapsaicin led to a concomitant downregulation of AP-1 activation. Further analysis of the molecular mechanism responsible for the dihydrocapsaicin-mediated decrease in phospho-p70S6K1, revealed that dihydrocapsaicin can block amino acid-dependent mechanistic targets of rapamycin complex 1 (mTORC1)-p70S6K1-S6 signal activation. Additionally, dihydrocapsaicin was able to selectively augment amino acid deprivation-induced cell death in mTORC1-hyperactive cells. Collectively, dihydrocapsaicin exerted chemopreventive effects through inhibiting amino acid signaling and c-Fos pathways and, thus, might be a promising cancer preventive natural agent.


Asunto(s)
Aminoácidos/metabolismo , Capsaicina/análogos & derivados , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Genes fos/fisiología , Animales , Capsaicina/química , Capsaicina/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genes fos/genética , Humanos , Ratones , Estructura Molecular , Fosforilación , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Acetato de Tetradecanoilforbol/metabolismo
19.
Behav Neurosci ; 133(5): 489-495, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31058522

RESUMEN

Cocaine-use disorders are characterized by repeated relapse to drug-seeking and drug-taking behavior following periods of abstinence. Former drug users display increased activation of the orbitofrontal cortex (OFC) in response to drug-related cues, and similar phenomena are also observed in rodent models of drug relapse. The lateral, but not medial, OFC functionally contributes to the maintenance of cue-drug associations; however, less is known about the role of the ventral OFC in this process. To examine the pattern of neuronal activation in OFC subregions in response to drug-associated cues, rats were trained to respond on a lever for a cocaine infusion paired with a complex cue (2-hr sessions, minimum 10 days). Cocaine self-administration was followed by extinction training, in which lever responses resulted in no consequences (2-hr sessions, minimum 7 days). During a 1-hr reinstatement test, drug-seeking behavior (i.e., responses on the drug-paired lever) was examined in the presence or absence of contingent drug-paired cues (Cue TEST vs. Ext TEST, respectively). Rats were overdosed with a ketamine + xylazine cocktail 30-min post session, and transcardially perfused with 4% paraformaldehyde. Cfos protein expression was utilized to measure potential changes in neural activation between the reinstatement test groups. An increase in the number of Cfos-Immunoreactive cells was observed in the ventral and lateral subregions of the OFC in the Cue TEST group. The present findings provide evidence that the ventral and lateral regions of the rat OFC display similar patterns of neuronal activation in response to cocaine-paired cues. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Asunto(s)
Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/fisiología , Corteza Prefrontal/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Cocaína/farmacología , Condicionamiento Operante/fisiología , Señales (Psicología) , Extinción Psicológica/efectos de los fármacos , Genes fos/genética , Masculino , Neuronas/efectos de los fármacos , Corteza Prefrontal/fisiología , Ratas , Ratas Sprague-Dawley , Autoadministración , Transcriptoma/genética
20.
Invest Ophthalmol Vis Sci ; 60(5): 1706-1713, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31013343

RESUMEN

Purpose: Ocular pain and discomfort are the most defining symptoms of dry eye disease. We determined the ability of topical progesterone to affect corneal sensitivity and brainstem processing of nociceptive inputs. Methods: Progesterone or vehicle gel was applied to the shaved forehead in male Sprague Dawley rats. As a site control, gel also was applied to the cheek on the side contralateral to corneal stimulation. Corneal mechanical thresholds were determined using the Cochet-Bonnet esthesiometer in intact and lacrimal gland excision-induced dry eye animals. Eye wipe behaviors in response to hypertonic saline and capsaicin were examined, and corneal mustard oil-induced c-Fos immunohistochemistry was quantified in the brainstem spinal trigeminal nucleus. Results: Progesterone gel application to the forehead, but not the contralateral cheek, increased corneal mechanical thresholds in intact and lacrimal gland excision animals beginning <30 minutes after treatment. Subcutaneous injection of the local anesthetic bupivacaine into the forehead region before application of progesterone prevented the increase in corneal mechanical thresholds. Furthermore, progesterone decreased capsaicin-evoked eye wipe behavior in intact animals and hypertonic saline evoked eye wipe behavior in dry eye animals. The number of Fos-positive neurons located in the caudal region of the spinal trigeminal nucleus after corneal mustard oil application was reduced in progesterone-treated animals. Conclusions: Results from this study indicate that progesterone, when applied to the forehead, produces analgesia as indicated by increased corneal mechanical thresholds and decreased nociceptive responses to hypertonic saline and capsaicin.


Asunto(s)
Analgésicos/administración & dosificación , Enfermedades de la Córnea/prevención & control , Síndromes de Ojo Seco/complicaciones , Dolor Ocular/prevención & control , Frente , Progesterona/administración & dosificación , Progestinas/administración & dosificación , Animales , Fenómenos Biomecánicos/fisiología , Capsaicina/administración & dosificación , Córnea/fisiopatología , Enfermedades de la Córnea/etiología , Enfermedades de la Córnea/fisiopatología , Modelos Animales de Enfermedad , Dolor Ocular/etiología , Dolor Ocular/fisiopatología , Regulación de la Expresión Génica/fisiología , Genes fos/genética , Masculino , Planta de la Mostaza , Neuronas/fisiología , Dimensión del Dolor , Aceites de Plantas/administración & dosificación , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA