Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
1.
Int J Mol Sci ; 22(20)2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34681812

RESUMEN

Growth and differentiation factor 15 (GDF15), a divergent member of the transforming growth factor-ß (TGF-ß) superfamily, has been reported to be overexpressed in different kinds of cancer types. However, the function and mechanism of GDF15 in head and neck cancer (HNC) remains unclear. The Cancer Genome Atlas (TCGA) data show that the expression of GDF15 is significantly associated with tumor AJCC stage, lymph vascular invasion and tumor grade in HNC. In this study, we confirmed that knockdown of GDF15 attenuated: cell proliferation, migration and invasion via regulation of EMT through a canonical pathway; SMAD2/3 and noncanonical pathways; PI3K/AKT and MEK/ERK in HNC cell lines. Furthermore, we found that early growth response 1 (EGR1) was a transcription factor of GDF15. Interestingly, we also demonstrated that GDF15 could regulate the expression of EGR1, which meant a positive feedback loop occurred between these two factors. Moreover, combined inhibition of both GDF15 and EGR1 in a HNC mouse xenograft model showed significantly decreased tumor volume compared to inhibition of EGR1 or GDF15 alone. Our study showed that the GDF15-EGR1 signaling axis may be a good target in HNC patients.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Factor 15 de Diferenciación de Crecimiento/genética , Neoplasias de Cabeza y Cuello/patología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Transición Epitelial-Mesenquimal/genética , Retroalimentación Fisiológica/fisiología , Regulación Neoplásica de la Expresión Génica , Factor 15 de Diferenciación de Crecimiento/fisiología , Células HaCaT , Neoplasias de Cabeza y Cuello/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal/genética , Células Tumorales Cultivadas
2.
Biol Res ; 54(1): 9, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712084

RESUMEN

BACKGROUND: PGF2α is essential for the induction of the corpus luteum regression which in turn reduces progesterone production. Early growth response (EGR) proteins are Cys2-His2-type zinc-finger transcription factor that are strongly linked to cellular proliferation, survival and apoptosis. Rapid elevation of EGR1 was observed after luteolytic dose of PGF2α. EGR1 is involved in the transactivation of many genes, including TGFß1, which plays an important role during luteal regression. METHODS: The current study was conducted in buffalo luteal cells with the aim to better understand the role of EGR1 in transactivation of TGFß1 during PGF2α induced luteal regression. Luteal cells from mid stage corpus luteum of buffalo were cultured and treated with different doses of PGF2α for different time durations. Relative expression of mRNAs encoding for enzymes within the progesterone biosynthetic pathway (3ßHSD, CYP11A1 and StAR); Caspase 3; AKT were analyzed to confirm the occurrence of luteolytic event. To determine if EGR1 is involved in the PGF2α induced luteal regression via induction of TGFß1 expression, we knocked out the EGR1 gene by using CRISPR/Cas9. RESULT: The present experiment determined whether EGR1 protein expression in luteal cells was responsive to PGF2α treatment. Quantification of EGR1 and TGFß1 mRNA showed significant up regulation in luteal cells of buffalo at 12 h post PGF2α induction. In order to validate the role of PGF2α on stimulating the expression of TGFß1 by an EGR1 dependent mechanism we knocked out EGR1. The EGR1 ablated luteal cells were stimulated with PGF2α and it was observed that EGR1 KO did not modulate the PGF2α induced expression of TGFß1. In PGF2α treated EGR1 KO luteal cell, the mRNA expression of Caspase 3 was significantly increased compared to PGF2α treated wild type luteal cells maintained for 12 h. We also studied the influence of EGR1 on steroidogenesis. The EGR1 KO luteal cells with PGF2α treatment showed no substantial difference either in the progesterone concentration or in StAR mRNA expression with PGF2α-treated wild type luteal cells. CONCLUSION: These results suggest that EGR1 signaling is not the only factor which plays a role in the regulation of PGF2α induced TGFß1 signaling for luteolysis.


Asunto(s)
Búfalos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Cuerpo Lúteo/fisiología , Dinoprost , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Luteólisis , Animales , Células Cultivadas , Cuerpo Lúteo/citología , Dinoprost/farmacología , Femenino , Regulación de la Expresión Génica , Transducción de Señal , Factor de Crecimiento Transformador beta1/fisiología
3.
Biol. Res ; 54: 9-9, 2021. ilus, tab, graf
Artículo en Inglés | LILACS | ID: biblio-1505802

RESUMEN

BACKGROUND: PGF2α is essential for the induction of the corpus luteum regression which in turn reduces progesterone production. Early growth response (EGR) proteins are Cys2-His2-type zinc-finger transcription factor that are strongly linked to cellular proliferation, survival and apoptosis. Rapid elevation of EGR1 was observed after luteolytic dose of PGF2α. EGR1 is involved in the transactivation of many genes, including TGFß1, which plays an important role during luteal regression. METHODS: The current study was conducted in buffalo luteal cells with the aim to better understand the role of EGR1 in transactivation of TGFß1 during PGF2α induced luteal regression. Luteal cells from mid stage corpus luteum of buffalo were cultured and treated with different doses of PGF2α for different time durations. Relative expression of mRNAs encoding for enzymes within the progesterone biosynthetic pathway (3ßHSD, CYP11A1 and StAR); Caspase 3; AKT were analyzed to confirm the occurrence of luteolytic event. To determine if EGR1 is involved in the PGF2α induced luteal regression via induction of TGFß1 expression, we knocked out the EGR1 gene by using CRISPR/Cas9. RESULT: The present experiment determined whether EGR1 protein expression in luteal cells was responsive to PGF2α treatment. Quantification of EGR1 and TGFß1 mRNA showed significant up regulation in luteal cells of buffalo at 12 h post PGF2α induction. In order to validate the role of PGF2α on stimulating the expression of TGFß1 by an EGR1 dependent mechanism we knocked out EGR1. The EGR1 ablated luteal cells were stimulated with PGF2α and it was observed that EGR1 KO did not modulate the PGF2α induced expression of TGFß1. In PGF2α treated EGR1 KO luteal cell, the mRNA expression of Caspase 3 was significantly increased compared to PGF2α treated wild type luteal cells maintained for 12 h. We also studied the influence of EGR1 on steroidogenesis. The EGR1 KO luteal cells with PGF2α treatment showed no substantial difference either in the progesterone concentration or in StAR mRNA expression with PGF2α-treated wild type luteal cells. CONCLUSION: These results suggest that EGR1 signaling is not the only factor which plays a role in the regulation of PGF2α induced TGFß1 signaling for luteolysis.


Asunto(s)
Animales , Femenino , Búfalos , Dinoprost/farmacología , Cuerpo Lúteo/fisiología , Luteólisis , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Transducción de Señal , Células Cultivadas , Regulación de la Expresión Génica , Cuerpo Lúteo/citología , Factor de Crecimiento Transformador beta1/fisiología
4.
Sci Rep ; 10(1): 15842, 2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32985557

RESUMEN

In mice, exercise, cold exposure and fasting lead to the differentiation of inducible-brown adipocytes, called beige adipocytes, within white adipose tissue and have beneficial effects on fat burning and metabolism, through heat production. This browning process is associated with an increased expression of the key thermogenic mitochondrial uncoupling protein 1, Ucp1. Egr1 transcription factor has been described as a regulator of white and beige differentiation programs, and Egr1 depletion is associated with a spontaneous increase of subcutaneous white adipose tissue browning, in absence of external stimulation. Here, we demonstrate that Egr1 mutant mice exhibit a restrained Ucp1 expression specifically increased in subcutaneous fat, resulting in a metabolic shift to a more brown-like, oxidative metabolism, which was not observed in other fat depots. In addition, Egr1 is necessary and sufficient to promote white and alter beige adipocyte differentiation of mouse stem cells. These results suggest that modulation of Egr1 expression could represent a promising therapeutic strategy to increase energy expenditure and to restrain obesity-associated metabolic disorders.


Asunto(s)
Adipocitos Beige/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Grasa Subcutánea/metabolismo , Adipocitos Beige/fisiología , Tejido Adiposo Blanco/fisiología , Animales , Diferenciación Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Femenino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Grasa Subcutánea/fisiología
5.
Mol Biol Rep ; 47(8): 5953-5962, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32705506

RESUMEN

Pro-opiomelanocortin (POMC) is a large precursor protein of and ß-endorphin. POMC expressed in keratinocytes regulates various pathophysiological responses, such as pruritus in atopic dermatitis. Interleukin (IL)-31 is a T helper 2 (Th2)-derived cytokine that functions as a pruritogen, stimulating the sensory neurons in the skin. However, the regulatory mechanism underlying IL-31-induced POMC expression in keratinocytes remains largely unknown. Herein, using a 5'-serial deletion and site-specific mutation constructs of the regulatory region of POMC, we demonstrated that a putative EGR1-binding sequence (EBS) motif in POMC is required for its upregulation by IL-31 in HaCaT keratinocytes. Notably, EGR-1 directly interacted with the EBS motif in POMC. The ectopic expression of EGR-1 stimulated the POMC promoter activity, whereas the knockdown of EGR-1 expression by RNA interference reduced IL-31-induced POMC expression. Furthermore, we observed that three major mitogen-activated protein kinases, ERK, JNK, and p38 kinase, mediated IL-31-induced EGR-1 expression. In summary, our results suggest that EGR-1 trans-activates POMC in response to IL-31 stimulation in HaCaT keratinocytes.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Interleucinas/farmacología , Queratinocitos/metabolismo , Proopiomelanocortina/genética , Transcripción Genética/efectos de los fármacos , Secuencias de Aminoácidos , Línea Celular Transformada , Proteína 1 de la Respuesta de Crecimiento Precoz/antagonistas & inhibidores , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Genes Reporteros , Genes Sintéticos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mutagénesis Sitio-Dirigida , Mutación Puntual , Proopiomelanocortina/biosíntesis , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Activación Transcripcional , Regulación hacia Arriba/efectos de los fármacos
6.
Arterioscler Thromb Vasc Biol ; 40(8): 1905-1917, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32580633

RESUMEN

OBJECTIVE: Remodeling of the extracellular matrix plays a vital role in cardiovascular diseases. Using a mouse model of postnatal ascending aortic aneurysms (termed Fbln4SMKO), we have reported that abnormal mechanosensing led to aneurysm formation in Fbln4SMKO with an upregulation of the mechanosensitive transcription factor, Egr1 (Early growth response 1). However, the role of Egr1 and its upstream regulator(s) in the initiation of aneurysm development and their relationship to an aneurysmal microenvironment are unknown. Approach and Results: To investigate the contribution of Egr1 in the aneurysm development, we deleted Egr1 in Fbln4SMKO mice and generated double knockout mice (DKO, Fbln4SMKO; Egr1-/-). Aneurysms were prevented in DKO mice (42.8%) and Fbln4SMKO; Egr1+/- mice (26%). Ingenuity Pathway Analysis identified PAR1 (protease-activated receptor 1) as a potential Egr1 upstream gene. Protein and transcript levels of PAR1 were highly increased in Fbln4SMKO aortas at postnatal day 1 before aneurysm formed, together with active thrombin and MMP (matrix metalloproteinase)-9, both of which serve as a PAR1 activator. Concordantly, protein levels of PAR1, Egr1, and thrombin were significantly increased in human thoracic aortic aneurysms. In vitro cyclic stretch assays (1.0 Hz, 20% strain, 8 hours) using mouse primary vascular smooth muscle cells induced marked expression of PAR1 and secretion of prothrombin in response to mechanical stretch. Thrombin was sufficient to induce Egr1 expression in a PAR1-dependent manner. CONCLUSIONS: We propose that thrombin, MMP-9, and mechanical stimuli in the Fbln4SMKO aorta activate PAR1, leading to the upregulation of Egr1 and initiation of ascending aortic aneurysms.


Asunto(s)
Aneurisma de la Aorta Torácica/etiología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Proteínas de la Matriz Extracelular/fisiología , Receptor PAR-1/fisiología , Anciano , Anciano de 80 o más Años , Animales , Proteínas de la Matriz Extracelular/deficiencia , Femenino , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Persona de Mediana Edad , Receptor PAR-1/antagonistas & inhibidores , Estrés Mecánico , Trombina/farmacología
7.
Theranostics ; 10(9): 4233-4249, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32226550

RESUMEN

Rationale: Subjects unable to sustain ß-cell compensation develop type 2 diabetes. Early growth response-1 protein (EGR-1), implicated in the regulation of cell differentiation, proliferation, and apoptosis, is induced by diverse metabolic challenges, such as glucose or other nutrients. Therefore, we hypothesized that deficiency of EGR-1 might influence ß-cell compensation in response to metabolic overload. Methods: Mice deficient in EGR-1 (Egr1-/-) were used to investigate the in vivo roles of EGR-1 in regulation of glucose homeostasis and beta-cell compensatory responses. Results: In response to a high-fat diet, Egr1-/- mice failed to secrete sufficient insulin to clear glucose, which was associated with lower insulin content and attenuated hypertrophic response of islets. High-fat feeding caused a dramatic impairment in glucose-stimulated insulin secretion and downregulated the expression of genes encoding glucose sensing proteins. The cells co-expressing both insulin and glucagon were dramatically upregulated in islets of high-fat-fed Egr1-/- mice. EGR-1-deficient islets failed to maintain the transcriptional network for ß-cell compensatory response. In human pancreatic tissues, EGR1 expression correlated with the expression of ß-cell compensatory genes in the non-diabetic group, but not in the diabetic group. Conclusion: These results suggest that EGR-1 couples the transcriptional network to compensation for the loss of ß-cell function and identity. Thus, our study highlights the early stress coupler EGR-1 as a critical factor in the development of pancreatic islet failure.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Línea Celular Tumoral , Glucagón/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
8.
Sci Rep ; 9(1): 16620, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31719567

RESUMEN

Object recognition memory (ORM) serves to distinguish familiar items from novel ones. Reconsolidation is the process by which active memories are updated. The hippocampus is engaged in ORM reconsolidation through a mechanism involving induction of long-term potentiation (LTP). The transcription factor Zif268 is essential for hippocampal LTP maintenance and has been frequently associated with memory processes. However, its possible involvement in ORM reconsolidation has not been determined conclusively. Using Zif268 antisense oligonucleotides in combination with behavioural, biochemical and electrophysiological tools in rats, we found that hippocampal Zif268 is necessary to update ORM through reconsolidation but not to retrieve it or keep it stored. Our results also suggest that knocking down hippocampal Zif268 during ORM reconsolidation deletes the active recognition memory trace.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Hipocampo/fisiología , Consolidación de la Memoria/fisiología , Reconocimiento en Psicología/fisiología , Animales , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Masculino , Aprendizaje por Laberinto , Ratas , Ratas Wistar
9.
Oncogene ; 38(35): 6241-6255, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31312026

RESUMEN

Early growth response-1 (EGR1) is a transcription factor correlated with prostate cancer (PC) progression in a variety of contexts. For example, EGR1 levels increase in response to suppressed androgen receptor signaling or loss of the tumor suppressor, PTEN. EGR1 has been shown to regulate genes influencing proliferation, apoptosis, immune cell activation, and matrix degradation, among others. Despite this, the impact of EGR1 on PC metastatic colonization is unclear. We demonstrate using a PC model (DU145/RasB1) of bone and brain metastasis that EGR1 expression regulates angiogenic and osteoclastogenic properties of metastases. We have shown previously that FN14 (TNFRSF12A) and downstream NF-κB signaling is required for metastasis in this model. Here we demonstrate that FN14 ligation also leads to NF-κB-independent, MEK-dependent EGR1 expression. EGR1-depletion in DU145/RasB1 cells reduced both the number and size of metastases but did not affect primary tumor growth. Decreased EGR1 expression led to reduced blood vessel density in brain and bone metastases as well as decreased osteolytic bone lesion area and reduced numbers of osteoclasts at the bone-tumor interface. TWEAK (TNFSF12) induced several EGR1-dependent angiogenic and osteoclastogenic factors (e.g., PDGFA, TGFB1, SPP1, IL6, IL8, and TGFA, among others). Consistent with this, in clinical samples of PC, the level of several genes encoding angiogenic/osteoclastogenic pathway effectors correlated with EGR1 levels. Thus, we show here that EGR1 has a direct effect on prostate cancer metastases. EGR1 regulates angiogenic and osteoclastogenic factors, informing the underlying signaling networks that impact autonomous and microenvironmental mechanisms of cancer metastases.


Asunto(s)
Adenocarcinoma/patología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Neovascularización Patológica/genética , Osteogénesis/genética , Neoplasias de la Próstata/patología , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/genética , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Neovascularización Patológica/patología , Células PC-3 , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/genética , Células RAW 264.7 , Transducción de Señal/genética , Células Tumorales Cultivadas , Microambiente Tumoral/genética
10.
Phytother Res ; 33(6): 1736-1747, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31006910

RESUMEN

Glioblastoma multiforme (GBM) is a highly aggressive type of brain tumour. Patients with GBM respond poorly to chemotherapy and have poor survival outcomes. Neuron-glial antigen 2 (NG2), also known as chondroitin sulphate proteoglycan 4 (CSPG4), has been shown to contribute to critical processes, such as cell survival, proliferation, and chemotherapy resistance, during glioma progression. In this study, we found that furanodienone (FUR), a diene-type sesquiterpene isolated from the rhizomes of Rhizoma curcumae, exhibited a potential cytotoxic effect on temozolomide (TMZ)-resistant GBM cells in vitro by inhibiting CSPG4 and related signalling pathways. Studies investigating the mechanism demonstrated that FUR suppressed CSPG4-Akt-ERK signalling, inflammatory responses, and cytokine levels but activated caspase-dependent pathways and mitochondrial dysfunction. Furthermore, an immunofluorescence assay and a dual-luciferase reporter assay revealed that inhibition of EGR1-mediated transcription might have contributed to the FUR-dependent blockade of CSPG4 signalling and glioma cell survival. These results established a link between FUR-induced CSPG4 inhibition and the suppression of EGR1-dependent transcription. Attenuation of ERK1/2 and cytokine signalling might have generated the EGR1-dependent negative feedback loop of the CSPG4 pathway during FUR-induced apoptosis. These findings suggested that FUR could be a therapeutic candidate for the treatment of malignant glioma via targeting CSPG4 signalling.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Furanos/farmacología , Glioblastoma/tratamiento farmacológico , Sesquiterpenos/farmacología , Temozolomida/uso terapéutico , Adulto , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Resistencia a Antineoplásicos/genética , Furanos/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/patología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sesquiterpenos/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Temozolomida/administración & dosificación , Transcripción Genética/efectos de los fármacos
11.
Brain Pathol ; 29(4): 502-512, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30511454

RESUMEN

Our previous studies showed that the transcription factor early growth response-1 (EGR1) may play a role in keeping the brain cholinergic function intact in the preclinical stages of Alzheimer's disease (AD). In order to elucidate the mechanisms involved, we first performed data mining on our previous microarray study on postmortem human prefrontal cortex (PFC) for the changes in the expression of EGR1 and acetylcholinesterase (AChE) and the relationship between them during the course of AD. The study contained 49 patients, ranging from non-demented controls (Braak stage 0) to late AD patients (Braak stage VI). We found EGR1-mRNA was high in early AD and decreased in late AD stages, while AChE-mRNA was stable in preclinical AD and slightly decreased in late AD stages. A significant positive correlation was found between the mRNA levels of these two molecules. In addition, we studied the relationship between EGR1 and AChE mRNA levels in the frontal cortex of 3-12-months old triple-transgenic AD (3xTg-AD) mice. EGR1- and AChE-mRNA were lower in 3xTg-AD mice compared with wild-type (WT) mice. A significant positive correlation between these two molecules was present in the entire group and in each age group of either WT or 3xTg-AD mice. Subsequently, AChE expression was determined following up- or down-regulating EGR1 in cell lines and the EGR1 levels were found to regulate AChE at both the mRNA and protein levels. Dual-luciferase assay and electrophoretic mobility shift assay in the EGR1-overexpressing cells were performed to determine the functionally effective binding sites of the EGR1 on the AChE gene promoter. We conclude that the EGR1 can upregulate AChE expression by a direct effect on its gene promoter, which may contribute significantly to the changes in cholinergic function in the course of AD. The 3xTg-AD mouse model only reflects later stage AD.


Asunto(s)
Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Acetilcolinesterasa/fisiología , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Lóbulo Frontal/patología , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo
12.
Mol Biol Rep ; 46(1): 317-324, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30417207

RESUMEN

Leptin is a cytokine that regulates energy metabolism. Leptin can promote breast cancer progression in obese women. However, the mechanism of regulation of leptin expression in breast cancer cells is unclear. Tumor necrosis factor-alpha (TNF-α) stimulated the transcription of the leptin gene. Using mutant promoter constructs, we demonstrated that the EGR1-binding motif in the proximal region of the leptin gene is required for leptin transcription by TNF-α. Forced expression of EGR1 stimulated leptin promoter activity, whereas silencing of EGR1 by RNA interference reduced TNF-α-induced leptin protein accumulation. The ERK1/2 pathway contributed to the expression of EGR1 and leptin by TNF-α. Our results suggest that EGR1 targets the leptin gene in response to TNF-α stimulation in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Leptina/fisiología , Sitios de Unión , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Leptina/genética , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/fisiología
13.
J Neurosci ; 39(6): 1109-1118, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30587543

RESUMEN

Memory reconsolidation is hypothesized to be a mechanism by which memories can be updated with new information. Such updating has previously been shown to weaken memory expression or change the nature of the memory. Here we demonstrate that retrieval-induced memory destabilization also allows that memory to be strengthened by additional learning. We show that for rodent contextual fear memories, this retrieval conditioning effect is observed only when conditioning occurs within a specific temporal window opened by retrieval. Moreover, it necessitates hippocampal protein degradation at the proteasome and engages hippocampal Zif268 protein expression, both of which are established mechanisms of memory destabilization-reconsolidation. We also demonstrate a conceptually analogous pattern of results in human visual paired-associate learning. Retrieval-relearning strengthens memory performance, again only when relearning occurs within the temporal window of memory reconsolidation. These findings link retrieval-mediated learning in humans to the reconsolidation literature, and have potential implications both for the understanding of endogenous memory gains and strategies to boost weakly learned memories.SIGNIFICANCE STATEMENT Memory reconsolidation allows existing memories to be updated with new information. Previous research has demonstrated that reconsolidation can be manipulated pharmacologically and behaviorally to impair problematic memories. In this article, we show that reconsolidation can also be exploited to strengthen memory. This is shown both in rats, in a fear memory setting, and in a human declarative memory setting. For both, the behavioral conditions necessary to observe the memory strengthening match those that are required to trigger memory reconsolidation. There are several behavioral approaches that have previously been shown convincingly to strengthen memory. The present demonstration that reconsolidation can underpin long-lasting memory improvements may both provide an underlying mechanism for such approaches and provide new strategies to boost memories.


Asunto(s)
Hipocampo/fisiología , Aprendizaje/fisiología , Consolidación de la Memoria/fisiología , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Miedo/psicología , Humanos , Masculino , Recuerdo Mental/fisiología , Aprendizaje por Asociación de Pares/fisiología , Ratas
14.
Cell Signal ; 55: 8-16, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30557604

RESUMEN

High level expression of lipocalin 2 (LCN2) usually indicates poor prognosis in esophageal squamous cell carcinoma (ESCC) and many other cancers. Our previous study showed LCN2 promotes migration and invasion of ESCC cells through a novel positive feedback loop. However, the key transcription activation protein (KTAP) in the loop had not yet been identified. In this study, we first predicted the most probable KTAPs by bioinformatic analysis. We then assessed the transcription regulatory regions in the human LCN2 gene by fusing deletions of its 5'-flanking region to a dual-luciferase reporter. We found that the region -720/-200 containing transcription factor 7-like 2 (TCF7L2) (-273/-209) and early growth response 1 (EGR1) (-710/-616) binding sites is crucial for LCN2 promoter activity. Chromatin immunoprecipitation (ChIP) experiments demonstrated that TCF7L2 and EGR1 bound directly to their binding sites within the LCN2 promoter as KTAPs. Mechanistically, overexpression of TCF7L2 and EGR1 increased endogenous LCN2 expression via the ERK signaling pathway. Treatment with recombinant human LCN2 protein enhanced activation of the ERK pathway to facilitate endogenous LCN2 expression, as well as increase the expression level of TCF7L2 and EGR1. Treatment with the MEK inhibitor U0126 inhibited the activation by TCF7L2 or EGR1 overexpression. Moreover, overexpression of TCF7L2 or EGR1 accelerated the migration and invasion of ESCC cells. A synergistic effect was observed between TCF7L2 and EGR1 in amplifying the induction of LCN2 and enhancing migration and invasion. Taken together, our study indicates that TCF7L2 and EGR1 are the KTAPs of LCN2, within a positive "LCN2 → MEK/ERK → LCN2" path, to promote the migration and invasion of ESCC cells. Based on their clinicopathological significance, LCN2 and its two expression regulators TCF7L2 and ERG1 might be therapeutic targets for ESCC.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Lipocalina 2/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/fisiología , Línea Celular Tumoral , Movimiento Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Regiones Promotoras Genéticas
15.
Biomed Pharmacother ; 108: 1282-1288, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30372829

RESUMEN

Keloid is a fibrous benign tumor of the skin caused by increased fibroblast proliferation and overproduction of extracellular matrix (ECM) in the dermis. Several miRNAs exhibit critical roles in regulating keloid development. This is study aimed to investigate the effects and mechanisms of miR-203 in keloid fibroblasts. The miR-203 expression was detected by qRT-PCR; The cell viability was measured by MTT assay; The cell proliferation was measured by BrdU assay; The cell invasion was measured by Transwell assay; The protein expression was detected by Western blot; The target relationship between miR-203 and mRNA was measured by dual-luciferase assay. We found that miR-203 was significantly downregulated in both keloid tissues and keloid fibroblasts from keloid patients. MiR-203 overexpression in vitro led to a significant decrease of proliferation, invasion, and ECM production in keloid fibroblasts, whereas miR-203 inhibition induced the opposite results. A dual-luciferase reporter assay identified early growth response 1 (EGR1) and fibroblast growth factor 2 (FGF2) as targets of miR-203. EGR1 and FGF2 were overexpressed in keloid fibroblasts and negatively regulated by miR-203. Furthermore, overexpression of EGR1 and FGF2 partially attenuated the suppressive effect of miR-203 on the proliferation, invasion, and ECM production of keloid fibroblasts. In conclusion, we demonstrated for the first time that miR-203 decreased the proliferation, invasion, and ECM production of keloid fibroblasts by repressing EGR1 and FGF2 expression, suggesting a potential role of miR-203 in preventing and treating keloids.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteínas de la Matriz Extracelular/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/genética , Queloide/patología , MicroARNs/fisiología , Regiones no Traducidas 3' , Adulto , Proliferación Celular , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Femenino , Factor 2 de Crecimiento de Fibroblastos/fisiología , Fibroblastos/fisiología , Humanos , Queloide/metabolismo , Masculino , Persona de Mediana Edad
16.
Transfusion ; 58(4): 1054-1064, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29399809

RESUMEN

BACKGROUND: The P1 /P2 phenotypic polymorphism is one of the earliest blood groups discovered in humans. These blood groups have been connected to different levels of expression of the A4GALT gene in P1 and P2 red blood cells; however, the detailed molecular genetic mechanism that leads to these two phenotypes has not been established. STUDY DESIGN AND METHODS: After our previous identification of an association between the single-nucleotide polymorphisms (SNPs) rs2143918 and rs5751348 in A4GALT gene and the P1 /P2 phenotype, we conduct a survey of transcription factors that might connect these SNPs with the differential expression of the P1 -A4GALT and P2 -A4GALT alleles. An in silico analysis of potential transcription factor binding motifs within the polymorphic SNPs rs2143918 and rs5751348 genomic regions was performed, and this was followed by reporter assays examining the candidate transcription factors, gene expression profiling, electrophoretic mobility shift assays, and P1 -A4GALT and P2 -A4GALT allelic expression analysis. RESULTS: The results revealed that the differential binding of transcription factor early growth response 1 to the SNP rs5751348 genomic region with the different genotypes in the A4GALT gene leads to differential activation of P1 -A4GALT and P2 -A4GALT expression. CONCLUSION: The present investigation, together with our previous study (Lai et al., Transfusion 2014;54:3222-31), have elucidated the molecular genetic details associated with the P1 /P2 blood groups.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Galactosiltransferasas/biosíntesis , Regulación de la Expresión Génica , Polimorfismo de Nucleótido Simple , Alelos , Simulación por Computador , Factores de Transcripción de la Respuesta de Crecimiento Precoz/fisiología , Ensayo de Cambio de Movilidad Electroforética , Galactosiltransferasas/genética , Perfilación de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Unión Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Transcripción Genética
17.
Cell Signal ; 45: 102-109, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29408223

RESUMEN

Egr-1 is known to function mainly as a tumor suppressor through direct regulation of multiple tumor suppressor genes. To determine the role of Egr-1 in breast tumors in vivo, we used mouse models of breast cancer induced by HER2/neu. We compared neu-overexpressing Egr-1 knockout mice (neu/Egr-1 KO) to neu-overexpressing Egr-1 wild type or heterozygote mice (neu/Egr-1 WT or neu/Egr-1 het) with regard to onset of tumor appearance and number of tumors per mouse. In addition, to examine the role of Egr-1 in vitro, we established neu/Egr-1 WT and KO tumor cell lines derived from breast tumors developed in each mouse. Egr-1 deletion delayed tumor development in vivo and decreased the rate of cell growth in vitro. These results suggest that Egr-1 plays an oncogenic role in HER2/neu-driven mammary tumorigenesis.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Neoplasias Mamarias Experimentales/genética , Animales , Neoplasias de la Mama/patología , Proliferación Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Femenino , Eliminación de Gen , Neoplasias Mamarias Experimentales/patología , Ratones Noqueados , Receptor ErbB-2/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
18.
Mol Biol Cell ; 29(3): 356-362, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212876

RESUMEN

Loss of peroxisome proliferator-activated receptor γ (PPARγ) has been found to contribute to pulmonary artery smooth muscle cell (PASMC) proliferation and pulmonary arterial remodeling therefore the development of pulmonary hypertension (PH). Yet, the molecular mechanisms underlying PPARγ reduction in PASMC remain poorly understood. Here, we demonstrated that leptin dose- and time-dependently inducued PPARγ down-regulation and proliferation of primary cultured rat PASMC, this was accompanied with the activation of extracellular regulated kinase1/2 (ERK1/2) signaling pathway and subsequent induction of early growth response-1 (Egr-1) expression. The presence of MEK inhibitors U0126 or PD98059, or prior silencing Egr-1 with small interfering RNA suppressed leptin-induced PPARγ reduction. In addition, activation of PPARγ by pioglitazone or targeting ERK1/2/Egr-1 suppressed leptin-induced PASMC proliferation. Taken together, our study indicates that ERK1/2 signaling pathway-mediated leptin-induced PPARγ reduction and PASMC proliferation through up-regulation of Egr-1 and suggests that targeting leptin/ERK1/2/Egr-1 pathway might have potential value in ameliorating vascular remodeling and benefit PH.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Miocitos del Músculo Liso/fisiología , Arteria Pulmonar/citología , Animales , Proliferación Celular , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Hipertensión Pulmonar/metabolismo , Leptina/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Miocitos del Músculo Liso/metabolismo , PPAR gamma/metabolismo , PPAR gamma/fisiología , Cultivo Primario de Células , Arteria Pulmonar/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Remodelación Vascular
19.
FASEB J ; 32(3): 1184-1195, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29092905

RESUMEN

The harmonized actions of ovarian E2 and progesterone (P4) regulate the proliferation and differentiation of uterine cells in a spatiotemporal manner. Imbalances between these hormones often lead to infertility and gynecologic diseases. Whereas numerous factors that are involved in P4 signaling have been identified, few local factors that mediate E2 actions in the uterus have been revealed. Here, we demonstrate that estrogen induces the transcription factor, early growth response 1 ( Egr1), to fine-tune its actions in uterine epithelial cells (ECs) that are responsible for uterine receptivity for embryo implantation. In the presence of exogenous gonadotrophins, ovulation, fertilization, and embryonic development normally occur in Egr1-/- mice, but these animals experience the complete failure of embryo implantation with reduced artificial decidualization. Although serum levels of E2 and P4 were comparable between Egr1+/+ and Egr1-/- mice on d 4 of pregnancy, aberrantly reduced levels of progesterone receptor in Egr1-/- uterine ECs caused enhanced E2 activity and impaired P4 response. Ultrastructural analyses revealed that Egr1-/- ECs are not fully able to provide proper uterine receptivity. Uterine mRNA landscapes in Egr1-/- mice revealed that EGR1 controls the expression of a subset of E2-regulated genes. In addition, P4 signaling was unable to modulate estrogen actions, including those that are involved in cell-cycle progression, in ECs that were deficient in EGR1. Furthermore, primary coculture of Egr1-/- ECs with Egr1+/+ stromal cells, and vice versa, supported the notion that Egr1 is required to modulate E2 actions on ECs to prepare the uterine environment for embryo implantation. In contrast to its role in ECs, loss of Egr1 in stroma significantly reduced stromal cell proliferation. Collectively, our results demonstrate that E2 induces EGR1 to streamline its actions for the preparation of uterine receptivity for embryo implantation in mice.-Kim, H.-R., Kim, Y. S., Yoon, J. A., Yang, S. C., Park, M., Seol, D.-W., Lyu, S. W., Jun, J. H., Lim, H. J., Lee, D. R., Song, H. Estrogen induces EGR1 to fine-tune its actions on uterine epithelium by controlling PR signaling for successful embryo implantation.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Desarrollo Embrionario/efectos de los fármacos , Epitelio/metabolismo , Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Receptores de Progesterona/metabolismo , Útero/metabolismo , Animales , Células Cultivadas , Implantación del Embrión/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/patología , Femenino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Embarazo , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Útero/efectos de los fármacos , Útero/patología
20.
Acta Biochim Biophys Sin (Shanghai) ; 49(11): 1015-1021, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29036495

RESUMEN

Previous studies have shown that the differentiation potential declines with the age of progenitor cells and is linked to altered levels of senescence markers. The purpose of this study was to test whether senescence marker p16 affects age-related tenogenic differentiation in tendon stem/progenitor cells (TSPCs). Young and aged TSPCs were isolated from young/healthy and aged/degenerated human Achilles tendons, respectively. Cellular aging and capacity for tenogenic differentiation were examined. The results showed that the tenogenic differentiation capacity of TSPCs significantly decreases with advancing age. TSPCs from elderly donors showed upregulation of senescence-associated ß-galactosidase and p16 and concurrently a decrease in Type I collagen concentration and in the expressions of tendon-related markers: Scx, Tnmd, Bgn, Dcn, Col1, and Col3. Overexpression of p16 significantly inhibited tenogenic differentiation of young TSPCs. Analysis of the mechanism revealed that this effect is mediated by microRNA-217 and its target EGR1. These results indicated that p16 inhibits tenogenic differentiation of TSPCs via microRNA signaling pathways, which may serve as a potential target for the prevention or treatment in the future.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , MicroARNs/fisiología , Transducción de Señal/fisiología , Células Madre/metabolismo , Tendones/citología , Adolescente , Adulto , Factores de Edad , Anciano , Diferenciación Celular , Humanos , Persona de Mediana Edad , Células Madre/citología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...