Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 506
Filtrar
1.
Behav Brain Res ; 416: 113540, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34419513

RESUMEN

Dopamine (DA) transmission is important in the regulation of mood and anxiety behaviors. However, how specific dopaminergic signaling pathways respond to anxiogenic stimuli as well as regulate behaviors remains unknown. To understand how DA regulates the animal behaviors under anxiety we performed retrograde labeling and c-Fos staining of midbrain DA neurons. Our c-Fos labeling results showed that DA neurons projected to nucleus accumbens (NAc) are activated in animals treated with the elevated plus-maze (EPM). Real-time measurement of DA release using fast scanning cyclic voltammetry (FSCV) in NAc of freely behaving mice showed that increased DA release and more DA transients in the close arms than the open arms in the EPM. Meanwhile, we also observed a reduction of DA level from the close arms to the open arms. Local infusion of DA D1 receptor antagonist, SCH23390 in the core of NAc, leads to an anxiolytic-like effect in the open-field and EPM. These anxiolytic effects were not observed in animals received D2 receptor antagonist sulpiride infusion in the core of NAc. Taken together, our results reveal a novel function of the mesolimbic DA pathway through the D1 receptor in the regulation of anxiety-like behaviors.


Asunto(s)
Ansiolíticos/farmacología , Benzazepinas/farmacología , Agonistas de Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Benzazepinas/antagonistas & inhibidores , Antagonistas de Dopamina/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Proteínas Proto-Oncogénicas c-fos/fisiología , Receptores de Dopamina D2/metabolismo , Transducción de Señal , Sulpirida/farmacología
2.
Sci Rep ; 11(1): 14677, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34282187

RESUMEN

Cancer stem-like cells (CSCs) have self-renewal abilities responsible for cancer progression, therapy resistance, and metastatic growth. The glioblastoma stem-like cells are the most studied among CSC populations. A recent study identified four transcription factors (SOX2, SALL2, OLIG2, and POU3F2) as the minimal core sufficient to reprogram differentiated glioblastoma (GBM) cells into stem-like cells. Transcriptomic data of GBM tissues and cell lines from two different datasets were then analyzed by the SWItch Miner (SWIM), a network-based software, and FOSL1 was identified as a putative regulator of the previously identified minimal core. Herein, we selected NTERA-2 and HEK293T cells to perform an in vitro study to investigate the role of FOSL1 in the reprogramming mechanisms. We transfected the two cell lines with a constitutive FOSL1 cDNA plasmid. We demonstrated that FOSL1 directly regulates the four transcription factors binding their promoter regions, is involved in the deregulation of several stemness markers, and reduces the cells' ability to generate aggregates increasing the extracellular matrix component FN1. Although further experiments are necessary, our data suggest that FOSL1 reprograms the stemness by regulating the core of the four transcription factors.


Asunto(s)
Reprogramación Celular/genética , Células Madre Neoplásicas/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Diferenciación Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Células HEK293 , Células HeLa , Humanos , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-fos/genética
3.
Endocrinology ; 162(9)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34171102

RESUMEN

FOS, a subunit of the activator protein-1 (AP-1) transcription factor, has been implicated in various cellular changes. In the human ovary, the expression of FOS and its heterodimeric binding partners JUN, JUNB, and JUND increases in periovulatory follicles. However, the specific role of the FOS/AP-1 remains elusive. The present study determined the regulatory mechanisms driving the expression of FOS and its partners and functions of FOS using primary human granulosa/lutein cells (hGLCs). Human chorionic gonadotropin (hCG) induced a biphasic increase in the expression of FOS, peaking at 1 to 3 hours and 12 hours. The levels of JUN proteins were also increased by hCG, with varying expression patterns. Coimmunoprecipitation analyses revealed that FOS is present as heterodimers with all JUN proteins. hCG immediately activated protein kinase A and p42/44MAPK signaling pathways, and inhibitors for these pathways abolished hCG-induced increases in the levels of FOS, JUN, and JUNB. To identify the genes regulated by FOS, high-throughput RNA sequencing was performed using hGLC treated with hCG ± T-5224 (FOS inhibitor). Sequencing data analysis revealed that FOS inhibition affects the expression of numerous genes, including a cluster of genes involved in the periovulatory process such as matrix remodeling, prostaglandin synthesis, glycolysis, and cholesterol biosynthesis. Quantitative PCR analysis verified hCG-induced, T-5224-regulated expression of a selection of genes involved in these processes. Consistently, hCG-induced increases in metabolic activities and cholesterol levels were suppressed by T-5224. This study unveiled potential downstream target genes of and a role for the FOS/AP-1 complex in metabolic changes and cholesterol biosynthesis in granulosa/lutein cells of human periovulatory follicles.


Asunto(s)
Colesterol/biosíntesis , Metabolismo Energético/genética , Células de la Granulosa/metabolismo , Proteínas Proto-Oncogénicas c-fos/fisiología , Células Cultivadas , Gonadotropina Coriónica/farmacología , Metabolismo Energético/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Humanos , Ovulación/efectos de los fármacos , Ovulación/genética , Ovulación/metabolismo , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factores de Tiempo , Factor de Transcripción AP-1/efectos de los fármacos , Factor de Transcripción AP-1/fisiología
4.
Cancer Lett ; 506: 23-34, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33652085

RESUMEN

The AP-1 member Fra-1 is overexpressed in TNBC and plays crucial roles in tumor progression and treatment resistance. In a previous large-scale screen, we identified PARP1 to be among 118 proteins that interact with endogenous chromatin-bound Fra-1 in TNBC cells. PARP1 inhibitor (olaparib) is currently in clinical use for treatment of BRCA-mutated TNBC breast cancer. Here, we demonstrate that the Fra-1-PARP1 interaction impacts the efficacy of olaparib treatment. We show that PARP1 interacts with and downregulates Fra-1, thereby reducing AP-1 transcriptional activity. Olaparib treatment, or silencing of PARP1, consequently, increases Fra-1 levels and enhances its transcriptional activity. Increased Fra-1 can have adverse effect, including treatment resistance. We also found that a large fraction of PARP1-regulated genes was dependent on Fra-1. We show that by inhibiting Fra-1/AP-1, non-BRCA-mutated TNBC cells can become sensitized to olaparib treatment. We identify that high PARP1 expression is indicative of a poor clinical outcome in breast cancer patients overall (P = 0.01), but not for HER-2 positive patients. In conclusion, by exploring the functionality of the Fra-1 and PARP1 interaction, we propose that targeting Fra-1 could serve as a combinatory therapeutic approach to improve olaparib treatment outcome for TNBC patients.


Asunto(s)
Poli(ADP-Ribosa) Polimerasa-1/fisiología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas Proto-Oncogénicas c-fos/fisiología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antígeno B7-H1/fisiología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ftalazinas/farmacología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Factor de Transcripción AP-1/fisiología
5.
Nucleic Acids Res ; 49(5): 2488-2508, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33533919

RESUMEN

The ubiquitous family of dimeric transcription factors AP-1 is made up of Fos and Jun family proteins. It has long been thought to operate principally at gene promoters and how it controls transcription is still ill-understood. The Fos family protein Fra-1 is overexpressed in triple negative breast cancers (TNBCs) where it contributes to tumor aggressiveness. To address its transcriptional actions in TNBCs, we combined transcriptomics, ChIP-seqs, machine learning and NG Capture-C. Additionally, we studied its Fos family kin Fra-2 also expressed in TNBCs, albeit much less. Consistently with their pleiotropic effects, Fra-1 and Fra-2 up- and downregulate individually, together or redundantly many genes associated with a wide range of biological processes. Target gene regulation is principally due to binding of Fra-1 and Fra-2 at regulatory elements located distantly from cognate promoters where Fra-1 modulates the recruitment of the transcriptional co-regulator p300/CBP and where differences in AP-1 variant motif recognition can underlie preferential Fra-1- or Fra-2 bindings. Our work also shows no major role for Fra-1 in chromatin architecture control at target gene loci, but suggests collaboration between Fra-1-bound and -unbound enhancers within chromatin hubs sometimes including promoters for other Fra-1-regulated genes. Our work impacts our view of AP-1.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Sitios de Unión , Línea Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Epigénesis Genética , Antígeno 2 Relacionado con Fos/metabolismo , Humanos , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/fisiología , Factor de Transcripción AP-1/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Factores de Transcripción p300-CBP/metabolismo
6.
Learn Mem ; 27(11): 467-476, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33060284

RESUMEN

The delayed match-to-sample task (DMS) is used to probe working memory (WM) across species. While the involvement of the PFC in this task has been established, limited information exists regarding the recruitment of broader circuitry, especially under the low- versus high-WM load. We sought to address this question by using a variable-delay operant DMS task. Male Sprague-Dawley rats were trained and tested to determine their baseline WM performance across all (0- to 24-sec) delays. Next, rats were tested in a single DMS test with either 0- or 24-sec fixed delay, to assess low-/high-load WM performance. c-Fos mRNA expression was quantified within cortical and subcortical regions and correlated with WM performance. High WM load up-regulated overall c-Fos mRNA expression within the PrL, as well as within a subset of mGlu5+ cells, with load-dependent, local activation of protein kinase C (PKC) as the proposed underlying molecular mechanism. The PrL activity negatively correlated with choice accuracy during high load WM performance. A broader circuitry, including several subcortical regions, was found to be activated under low and/or high load conditions. These findings highlight the role of mGlu5- and/or PKC-dependent signaling within the PrL, and corresponding recruitment of subcortical regions during high-load WM performance.


Asunto(s)
Condicionamiento Operante , Memoria a Corto Plazo , Animales , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Condicionamiento Operante/fisiología , Masculino , Memoria a Corto Plazo/fisiología , Proteína Quinasa C/metabolismo , Proteína Quinasa C/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-fos/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Nat Commun ; 11(1): 4484, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32901027

RESUMEN

Chronic stress is a key risk factor for mood disorders like depression, but the stress-induced changes in brain circuit function and gene expression underlying depression symptoms are not completely understood, hindering development of novel treatments. Because of its projections to brain regions regulating reward and anxiety, the ventral hippocampus is uniquely poised to translate the experience of stress into altered brain function and pathological mood, though the cellular and molecular mechanisms of this process are not fully understood. Here, we use a novel method of circuit-specific gene editing to show that the transcription factor ΔFosB drives projection-specific activity of ventral hippocampus glutamatergic neurons causing behaviorally diverse responses to stress. We establish molecular, cellular, and circuit-level mechanisms for depression- and anxiety-like behavior in response to stress and use circuit-specific gene expression profiling to uncover novel downstream targets as potential sites of therapeutic intervention in depression.


Asunto(s)
Reacción de Prevención/fisiología , Hipocampo/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Animales , Ansiedad/metabolismo , Conducta Animal/fisiología , Técnicas de Inactivación de Genes , Silenciador del Gen , Hipocampo/anatomía & histología , Hipocampo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/deficiencia , Proteínas Proto-Oncogénicas c-fos/genética , Conducta Social , Estrés Psicológico
8.
Oncogene ; 39(22): 4436-4449, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32350443

RESUMEN

Activator protein (AP)-1 transcription factors are essential elements of the pro-oncogenic functions of transforming growth factor-ß (TGFß)-SMAD signaling. Here we show that in multiple HER2+ and/or EGFR+ breast cancer cell lines these AP-1-dependent tumorigenic properties of TGFß critically rely on epidermal growth factor receptor (EGFR) activation and expression of the ΔN isoform of transcriptional regulator p63. EGFR and ΔNp63 enabled and/or potentiated the activation of a subset of TGFß-inducible invasion/migration-associated genes, e.g., ITGA2, LAMB3, and WNT7A/B, and enhanced the recruitment of SMAD2/3 to these genes. The TGFß- and EGF-induced binding of SMAD2/3 and JUNB to these gene loci was accompanied by p63-SMAD2/3 and p63-JUNB complex formation. p63 and EGFR were also found to strongly potentiate TGFß induction of AP-1 proteins and, in particular, FOS family members. Ectopic overexpression of FOS could counteract the decrease in TGFß-induced gene activation after p63 depletion. p63 is also involved in the transcriptional regulation of heparin binding (HB)-EGF and EGFR genes, thereby establishing a self-amplification loop that facilitates and empowers the pro-invasive functions of TGFß. These cooperative pro-oncogenic functions of EGFR, AP-1, p63, and TGFß were efficiently inhibited by clinically relevant chemical inhibitors. Our findings may, therefore, be of importance for therapy of patients with breast cancers with an activated EGFR-RAS-RAF pathway.


Asunto(s)
Neoplasias de la Mama/patología , Factor de Crecimiento Epidérmico/fisiología , Regulación Neoplásica de la Expresión Génica , Invasividad Neoplásica/genética , Proteínas de Neoplasias/fisiología , Transducción de Señal , Factor de Transcripción AP-1/genética , Factores de Transcripción/genética , Transcripción Genética , Factor de Crecimiento Transformador beta1/fisiología , Proteínas Supresoras de Tumor/genética , Neoplasias de la Mama/química , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Receptores ErbB/fisiología , Femenino , Humanos , Sistema de Señalización de MAP Quinasas , Proteínas de Neoplasias/genética , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Receptor ErbB-2/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta/fisiología , Proteínas Smad/fisiología
9.
Nat Neurosci ; 23(6): 707-717, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32451484

RESUMEN

Neuronal activation induces rapid transcription of immediate early genes (IEGs) and longer-term chromatin remodeling around secondary response genes (SRGs). Here, we use high-resolution chromosome-conformation-capture carbon-copy sequencing (5C-seq) to elucidate the extent to which long-range chromatin loops are altered during short- and long-term changes in neural activity. We find that more than 10% of loops surrounding select IEGs, SRGs, and synaptic genes are induced de novo during cortical neuron activation. IEGs Fos and Arc connect to activity-dependent enhancers via singular short-range loops that form within 20 min after stimulation, prior to peak messenger RNA levels. By contrast, the SRG Bdnf engages in both pre-existing and activity-inducible loops that form within 1-6 h. We also show that common single-nucleotide variants that are associated with autism and schizophrenia are colocalized with distinct classes of activity-dependent, looped enhancers. Our data link architectural complexity to transcriptional kinetics and reveal the rapid timescale by which higher-order chromatin architecture reconfigures during neuronal stimulation.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Expresión Génica/fisiología , Genoma/genética , Neuronas/fisiología , Animales , Bicuculina/farmacología , Factor Neurotrófico Derivado del Encéfalo/fisiología , Ensamble y Desensamble de Cromatina/genética , Proteínas del Citoesqueleto/fisiología , Genoma/efectos de los fármacos , Humanos , Ratones , Proteínas del Tejido Nervioso/fisiología , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/fisiología , Tetrodotoxina/farmacología , Factores de Tiempo
10.
Neurobiol Aging ; 87: 98-107, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31889558

RESUMEN

Homing pigeons experience age-related spatial-cognitive decline similar to that seen in mammals. In contrast to mammals, however, previous studies have shown the hippocampal formation (HF) of old, cognitively impaired pigeons to be greater in volume and neuron number compared with young pigeons. As a partial explanation of the cognitive decline in older birds, it was hypothesized that older pigeons have reduced HF activation during spatial learning. The present study compared HF activation (via the activity-dependent expression of the immediate early gene c-Fos) between younger and older pigeons during learning of a spatial, delayed nonmatch-to-sample task. On the last day of training, c-Fos activation significantly correlated with behavioral performance in the young, but not old, pigeons suggesting more HF engagement by the young pigeons in solving the task. The behavioral correlation was additionally associated with consistently higher, but insignificant c-Fos activation across practically every HF subdivision in the young compared with the old pigeons. In sum, the results of the present study are consistent with the hypothesis that age-related decline in the spatial cognitive ability of homing pigeons is in part a result of an older HF being less responsive to the processing of spatial information. However, alternative interpretations of the data are discussed.


Asunto(s)
Envejecimiento/genética , Envejecimiento/fisiología , Conducta Animal , Envejecimiento Cognitivo/psicología , Columbidae/genética , Columbidae/fisiología , Hipocampo/patología , Hipocampo/fisiopatología , Fenómenos de Retorno al Lugar Habitual , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Memoria Espacial/fisiología , Animales , Expresión Génica , Neuronas/patología , Tamaño de los Órganos , Proteínas Proto-Oncogénicas c-fos/fisiología
11.
eNeuro ; 6(3)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31138660

RESUMEN

Acquisition of social proficiency entails behavioral adaptations to social experience, including both behavioral flexibility and inhibition of behaviors inappropriate in specific social contexts. Here, we investigated the contributions of testosterone and ΔFosB, a transcription factor linked to experience-dependent neural plasticity, to the adolescent maturation of social proficiency in male-female social interactions. To determine whether pubertal testosterone organizes circuits underlying social proficiency, we first compared behavioral adaptations to sexual experience in male Syrian hamsters that were deprived of testosterone during puberty (prepubertal castration; NoT@P) to those of males deprived of testosterone for an equivalent period of time in adulthood (postpubertal castration; T@P). All males were given testosterone replacement in adulthood for two weeks before sexual behavior testing, where males were allowed to interact with a receptive female once per week for five consecutive weeks. T@P males showed the expected decrease in ectopic (mis-directed) mounts with sexual experience, whereas NoT@P males did not. In addition, sexual experience induced FosB gene products expression in the infralimbic cortex (IL) in T@P, but not NoT@P, males. Overexpression of ΔFosB via an adeno-associated viral (AAV) vector in the IL of NoT@P males prior to sexual behavior testing was sufficient to produce a behavioral phenotype similar to that of experienced T@P males. Finally, overexpression of ΔFosB in IL increased the density of immature spines on IL dendrites. Our findings provide evidence that social proficiency acquired through sexual experience is organized by pubertal testosterone through the regulation of ΔFosB in the IL, possibly through increasing synaptic lability.


Asunto(s)
Mesocricetus/fisiología , Corteza Prefrontal/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Pubertad/fisiología , Pubertad/psicología , Conducta Sexual Animal/fisiología , Testosterona/fisiología , Adaptación Fisiológica , Animales , Femenino , Relaciones Interpersonales , Masculino
12.
Behav Neurosci ; 133(4): 378-384, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30869949

RESUMEN

Motivated behaviors share the common feature of activating the mesolimbic dopamine system. Repeated experience with motivated behaviors can cause long-lasting structural changes in the nucleus accumbens (NAc). The molecular mechanisms underlying this experience-dependent plasticity in the NAc have been well described following experience with drugs of abuse. In particular, the transcription factor Delta FosB (ΔFosB) is a key regulator of drug-related neuroplasticity. Fewer studies have examined the molecular mechanisms underlying experience-dependent plasticity in the NAc following naturally motivated behaviors, but previous research has demonstrated that sexual experience increases the accumulation of ΔFosB in the NAc of female hamsters and male rats. Sex behavior is unique among motivated behaviors in that the expression of the behavior varies drastically between males and females of the same species. Despite this, a quantitative comparison of ΔFosB following sex experience in males and females of the same species has never been conducted. We therefore used Western blotting to test the hypothesis that sex experience increases ΔFosB in both male and female Syrian hamsters following repeated sexual experience. We found that sex experience significantly increases ΔFosB protein in male and female Syrian hamsters. Further, ΔFosB protein levels did not differ between males and females following sex experience. Interestingly, repeated sex experience only led to increased copulatory efficiency in female hamsters; male copulatory efficiency did not improve with repeated experience. Together, these data demonstrate that ΔFosB is increased following sexual reward in both males and females but may be uncoupled from behavioral plasticity in males. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Asunto(s)
Motivación/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Conducta Sexual Animal/fisiología , Animales , Encéfalo/fisiología , Copulación/fisiología , Cricetinae , Femenino , Masculino , Mesocricetus/metabolismo , Plasticidad Neuronal/fisiología , Núcleo Accumbens/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Recompensa , Factores Sexuales
13.
FASEB J ; 33(6): 7603-7614, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30892941

RESUMEN

Vascular aging has a strong relationship with cardiovascular disease. Fos-related antigen 1 (Fra-1), also referred to as Fos-like antigen 1, is a transcription factor and has been reported to be involved in many pathologic processes. Here, we demonstrate that Fra-1 plays a critical role in angiotensin II (Ang II)-induced vascular senescence. Fra-1 expression is increased significantly in Ang II-induced rat aortic endothelial cell (RAEC) senescence and the arteries from Ang II-infused mice. Interestingly, silencing Fra-1 blocks Ang II-induced senescence phenotypes in RAECs, including decreased senescence-associated ß-galactosidase staining, and mitigated proliferation suppression and senescence-associated secretory phenotype. Further, knocking down Fra-1 inhibits vascular aging phenotypes in an Ang II-infused mice model. The up-regulated Fra-1 also exists in human atherosclerotic plaques and Ang II-induced vascular smooth muscle cells as well as in replicated senescence RAECs. Mechanistic studies reveal that Fra-1 preferentially associates with c-Jun and binds to the cyclin-dependent kinase inhibitor 1a (p21) and cyclin-dependent kinase inhibitor 2a (p16) promoter region, leading to elevated gene expression, which causes senescence-related phenotypes. In conclusion, our results identify that Fra-1 plays a novel and key role in promoting vascular aging by directly binding and transcriptionally activating p21 and p16 signaling, suggesting intervention of Fra-1 is a potential strategy for preventing aging-associated cardiovascular disorders.-Yang, D., Xiao, C., Long, F., Wu, W., Huang, M., Qu, L., Liu, X., Zhu, Y. Fra-1 plays a critical role in angiotensin II-induced vascular senescence.


Asunto(s)
Angiotensina II/fisiología , Músculo Liso Vascular/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Animales , Células Cultivadas , Senescencia Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Genes jun , Genes p16 , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Transducción de Señal
14.
Eur Rev Med Pharmacol Sci ; 22(24): 8609-8615, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30575900

RESUMEN

OBJECTIVE: To investigate the effect of highly-expressed FOSL1 on the tumorigenesis and metastasis of prostate cancer. PATIENTS AND METHODS: Researches were carried out in human prostate cancer tissues and cell lines. In prostate cancer tissues, the expression of FOSL was detected by immunohistochemistry. In vitro cell line experiments, we constructed a prostate cancer cell model with FOSL1 stable knockdown and tested cell proliferation and metastasis before and after knockdown of FOSL1. Finally, the epithelial-mesenchymal transition (EMT) markers before and after interference of FOSL1 were also analyzed. RESULTS: FOSL1 was confirmed to have a high expression in prostate cancer. Transwell experiments demonstrated that FOSL1 could enhance prostate cancer metastasis, while in vivo experiments revealed an accelerated progression of prostate cancer caused by FOSL1. In addition, Western blot analysis revealed an elevated level of N-cadherin and Snail1 and a reduced level of E-cadherin that was induced by FOSL1. CONCLUSIONS: FOSL1 can promote the occurrence and progression of prostate cancer by altering the EMT process of cells.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-fos/fisiología , Antígenos CD/análisis , Cadherinas/análisis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Masculino , Invasividad Neoplásica , Metástasis de la Neoplasia
15.
eNeuro ; 5(4)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30079375

RESUMEN

Both the function of hippocampal neurons and hippocampus-dependent behaviors are dependent on changes in gene expression, but the specific mechanisms that regulate gene expression in hippocampus are not yet fully understood. The stable, activity-dependent transcription factor ΔFosB plays a role in various forms of hippocampal-dependent learning and in the structural plasticity of synapses onto CA1 neurons. The authors examined the consequences of viral-mediated overexpression or inhibition of ΔFosB on the function of adult mouse hippocampal CA1 neurons using ex vivo slice whole-cell physiology. We found that the overexpression of ΔFosB decreased the excitability of CA1 pyramidal neurons, while inhibition increased excitability. Interestingly, these manipulations did not affect resting membrane potential or spike frequency adaptation, but ΔFosB overexpression reduced hyperpolarization-activated current. Both ΔFosB overexpression and inhibition decreased spontaneous excitatory postsynaptic currents, while only ΔFosB inhibition affected the AMPA/NMDA ratio, which was mediated by decreased NMDA receptor current, suggesting complex effects on synaptic inputs to CA1 that may be driven by homeostatic cell-autonomous or network-driven adaptations to the changes in CA1 cell excitability. Because ΔFosB is induced in hippocampus by drugs of abuse, stress, or antidepressant treatment, these results suggest that ΔFosB-driven changes in hippocampal cell excitability may be critical for learning and, in maladaptive states, are key drivers of aberrant hippocampal function in diseases such as addiction and depression.


Asunto(s)
Región CA1 Hipocampal/fisiología , Expresión Génica/fisiología , Aprendizaje/fisiología , Potenciales de la Membrana/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Células Piramidales/fisiología , Animales , Región CA1 Hipocampal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-fos/metabolismo , Células Piramidales/metabolismo
16.
Hear Res ; 366: 38-49, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29983289

RESUMEN

When a social sound category initially gains behavioral significance to an animal, plasticity events presumably enhance the ability to recognize that sound category in the future. In the context of learning natural social stimuli, neuromodulators such as norepinephrine and estrogen have been associated with experience-dependent plasticity and processing of newly salient social cues, yet continued plasticity once stimuli are familiar could disrupt the stability of sensorineural representations. Here we employed a maternal mouse model of natural sensory cortical plasticity for infant vocalizations to ask whether the engagement of the noradrenergic locus coeruleus (LC) by the playback of pup-calls is affected by either prior experience with the sounds or estrogen availability, using a well-studied cellular activity and plasticity marker, the immediate early gene c-Fos. We counted call-induced c-Fos immunoreactive (c-Fos-IR) cells in both LC and physiologically validated fields within the auditory cortex (AC) of estradiol or blank-implanted virgin female mice with either 0 or 5-days prior experience caring for vocalizing pups. Estradiol and pup experience interacted both in the induction of c-Fos-IR in the LC, as well as in behavioral measures of locomotion during playback, consistent with the neuromodulatory center's activity being an online reflection of both hormonal and experience-dependent influences on arousal. Throughout core AC, as well as in a high frequency sub-region of AC and in secondary AC, a main effect of pup experience was to reduce call-induced c-Fos-IR, irrespective of estradiol availability. This is consistent with the hypothesis that sound familiarity leads to less c-Fos-mediated plasticity, and less disrupted sensory representations of a meaningful call category. Taken together, our data support the view that any coupling between these sensory and neuromodulatory areas is situationally dependent, and their engagement depends differentially on both internal state factors like hormones and external state factors like prior experience.


Asunto(s)
Corteza Auditiva/fisiología , Estradiol/fisiología , Locus Coeruleus/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Estimulación Acústica , Animales , Corteza Auditiva/anatomía & histología , Conducta Animal/fisiología , Femenino , Inmunohistoquímica , Aprendizaje/fisiología , Locus Coeruleus/anatomía & histología , Ratones , Ratones Endogámicos CBA , Plasticidad Neuronal/fisiología , Norepinefrina/fisiología , Reconocimiento en Psicología/fisiología , Conducta Social , Vocalización Animal/fisiología
17.
Gastroenterology ; 155(3): 892-908.e6, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29864419

RESUMEN

BACKGROUND & AIMS: Cigarette smoking is a major risk factor for pancreatic cancer. Aggressive pancreatic tumors contain cancer cells with stem cell features. We investigated whether cigarette smoke induces stem cell features in pancreatic cancer cells. METHODS: KrasG12D; Pdx1-Cre mice were exposed to cigarette smoke or clean air (controls) for up to 20 weeks; pancreata were collected and analyzed by histology, quantitative reverse transcription polymerase chain reaction, and confocal immunofluorescence microscopy. HPNE and Capan1 cells were exposed to cigarette smoke extract (CSE), nicotine and nicotine-derived carcinogens (NNN or NNK), or clean air (controls) for 80 days and evaluated for stem cell markers and features using flow cytometry-based autofluorescence, sphere formation, and immunoblot assays. Proteins were knocked down in cells with small interfering RNAs. We performed RNA sequencing analyses of CSE-exposed cells. We used chromatin immunoprecipitation assays to confirm the binding of FOS-like 1, AP-1 transcription factor subunit (FOSL1) to RNA polymerase II-associated factor (PAF1) promoter. We obtained pancreatic ductal adenocarcinoma (PDAC) and matched nontumor tissues (n = 15) and performed immunohistochemical analyses. RESULTS: Chronic exposure of HPNE and Capan1 cells to CSE caused them to increase markers of stem cells, including autofluorescence and sphere formation, compared with control cells. These cells increased expression of ABCG2, SOX9, and PAF1, via cholinergic receptor nicotinic alpha 7 subunit (CHRNA7) signaling to mitogen-activated protein kinase 1 and FOSL1. CSE-exposed pancreatic cells with knockdown of PAF1 did not show stem cell features. Exposure of cells to NNN and NNK led to increased expression of CHRNA7, FOSL1, and PAF1 along with stem cell features. Pancreata from KrasG12D; Pdx1-Cre mice exposed to cigarette smoke had increased levels of PAF1 mRNA and protein, compared with control mice, as well as increased expression of SOX9. Levels of PAF1 and FOSL1 were increased in PDAC tissues, especially those from smokers, compared with nontumor pancreatic tissue. CSE exposure increased expression of PHD-finger protein 5A, a pluripotent transcription factor and its interaction with PAF1. CONCLUSIONS: Exposure to cigarette smoke activates stem cell features of pancreatic cells, via CHRNA7 signaling and FOSL1 activation of PAF1 expression. Levels of PAF1 are increased in pancreatic tumors of humans and mice with chronic cigarette smoke exposure.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Proteínas Portadoras/metabolismo , Fumar Cigarrillos/efectos adversos , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Carcinoma Ductal Pancreático/etiología , Línea Celular Tumoral , Humanos , Ratones , Páncreas/citología , Neoplasias Pancreáticas/etiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Transducción de Señal/fisiología , Receptor Nicotínico de Acetilcolina alfa 7/fisiología
18.
Oncol Rep ; 39(3): 1112-1118, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29399696

RESUMEN

Fos­related antigen 1 (Fra­1) has roles in a variety of cell functions, including cell proliferation, differentiation, transformation, and invasiveness, and it is upregulated in various cancers. We investigated the role of Fra­1 in cellular radioresistance using cells of two human colorectal cancer cell lines, SW620 and SW480. We found that SW620 cells are more sensitive than SW480 cells at doses greater than 6 Gy for X­ray or 3 Gy for carbon­ion (C­ion) radiation. Fra­1 expression tended to be decreased by the radiation in a dose­dependent manner in both cell lines; of note, a greater reduction of Fra­1 expression was observed in SW620 cells, especially at 6 Gy of X­ray or 3 Gy of C­ion irradiation, than in SW480 cells, indicating a possible association between Fra­1 downregulation and cellular radiosensitivity. Knockdown of Fra­1 in SW480 cells significantly increased the radiosensitivity to X­ray or C­ion radiation. On the other hand, overexpression of Fra­1 in SW620 cells significantly enhanced the radioresistance to C­ion radiation, suggesting a role of Fra­1 in radioresistance. Furthermore, we found that downregulation of Fra­1 protein in irradiated SW620 cells was regulated via protein degradation through a proteasome­dependent pathway. Overall, our results indicate a role of Fra­1 in radioresistance to both X­ray and C­ion radiation for colorectal cancer cell lines.


Asunto(s)
Neoplasias del Colon/metabolismo , Proteínas Proto-Oncogénicas c-fos/fisiología , Tolerancia a Radiación , Línea Celular Tumoral , Neoplasias del Colon/enzimología , Neoplasias del Colon/radioterapia , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Radiación Ionizante , Rayos X
19.
Mol Cell ; 68(6): 1067-1082.e12, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29272704

RESUMEN

Enhancer elements are genomic regulatory sequences that direct the selective expression of genes so that genetically identical cells can differentiate and acquire the highly specialized forms and functions required to build a functioning animal. To differentiate, cells must select from among the ∼106 enhancers encoded in the genome the thousands of enhancers that drive the gene programs that impart their distinct features. We used a genetic approach to identify transcription factors (TFs) required for enhancer selection in fibroblasts. This revealed that the broadly expressed, growth-factor-inducible TFs FOS/JUN (AP-1) play a central role in enhancer selection. FOS/JUN selects enhancers together with cell-type-specific TFs by collaboratively binding to nucleosomal enhancers and recruiting the SWI/SNF (BAF) chromatin remodeling complex to establish accessible chromatin. These experiments demonstrate how environmental signals acting via FOS/JUN and BAF coordinate with cell-type-specific TFs to select enhancer repertoires that enable differentiation during development.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Elementos de Facilitación Genéticos , Proteínas Proto-Oncogénicas c-fos/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Animales , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nucleosomas , Regiones Promotoras Genéticas , Factores de Transcripción/genética
20.
J Biol Chem ; 292(52): 21282-21290, 2017 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-29150442

RESUMEN

Epithelioid hemangioma is a locally aggressive vascular neoplasm, found in bones and soft tissue, whose cause is currently unknown, but may involve oncogene activation. FOS is one of the earliest viral oncogenes to be characterized, and normal cellular FOS forms part of the activator protein 1 (AP-1) transcription factor complex, which plays a pivotal role in cell growth, differentiation, and survival as well as the DNA damage response. Despite this, a causal link between aberrant FOS function and naturally occurring tumors has not yet been established. Here, we describe a thorough molecular and biochemical analysis of a mutant FOS protein we identified in these vascular tumors. The mutant protein lacks a highly conserved helix consisting of the C-terminal four amino acids of FOS, which we show is indispensable for fast, ubiquitin-independent FOS degradation via the 20S proteasome. Our work reveals that FOS stimulates endothelial sprouting and that perturbation of normal FOS degradation could account for the abnormal vessel growth typical of epithelioid hemangioma. To the best of our knowledge, this is the first functional characterization of mutant FOS proteins found in tumors.


Asunto(s)
Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/fisiología , Neoplasias Vasculares/genética , Inductores de la Angiogénesis , Carcinogénesis/genética , Carcinogénesis/metabolismo , Diferenciación Celular , Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes fos/genética , Hemangioma/genética , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Elementos Reguladores de la Transcripción/genética , Neoplasias Vasculares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA