Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biomed Sci ; 31(1): 10, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38243273

RESUMEN

BACKGROUND: The tumor microenvironment is characterized by inflammation-like and immunosuppression situations. Although cancer-associated fibroblasts (CAFs) are among the major stromal cell types in various solid cancers, including colon cancer, the interactions between CAFs and immune cells remains largely uncharacterized. Pentraxin 3 (PTX3) is responsive to proinflammatory cytokines and modulates immunity and tissue remodeling, but its involvement in tumor progression appears to be context-dependent and is unclear. METHODS: Open-access databases were utilized to examine the association of PTX3 expression and the fibroblast signature in colon cancer. Loss-of-function assays, including studies in tamoxifen-induced Ptx3 knockout mice and treatment with an anti-PTX3 neutralizing antibody (WHC-001), were conducted to assess the involvement of PTX3 in colon cancer progression as well as its immunosuppressive effect. Finally, bioinformatic analyses and in vitro assays were performed to reveal the downstream effectors and decipher the involvement of the CREB1/CEBPB axis in response to PTX3 and PTX3-induced promotion of M2 macrophage polarization. RESULTS: Clinically, higher PTX3 expression was positively correlated with fibroblasts and inflammatory response signatures and associated with a poor survival outcome in colon cancer patients. Blockade of PTX3 significantly reduced stromal cell-mediated tumor development. The decrease of the M2 macrophage population and an increase of the cytotoxic CD8+ T-cell population were observed following PTX3 inactivation in allografted colon tumors. We further revealed that activation of cyclic AMP-responsive element-binding protein 1 (CREB1) mediated the PTX3-induced promotion of M2 macrophage polarization. CONCLUSIONS: PTX3 contributes to stromal cell-mediated protumor immunity by increasing M2-like macrophage polarization, and inhibition of PTX3 with WHC-001 is a potential therapeutic strategy for colon cancer.


Asunto(s)
Neoplasias del Colon , Macrófagos , Componente Amiloide P Sérico , Animales , Ratones , Humanos , Macrófagos/metabolismo , Proteína C-Reactiva/genética , Neoplasias del Colon/genética , Terapia de Inmunosupresión , Microambiente Tumoral
2.
Mol Cell ; 48(5): 747-59, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23123197

RESUMEN

NPGPx is a member of the glutathione peroxidase (GPx) family; however, it lacks GPx enzymatic activity due to the absence of a critical selenocysteine residue, rendering its function an enigma. Here, we show that NPGPx is a newly identified stress sensor that transmits oxidative stress signals by forming the disulfide bond between its Cys57 and Cys86 residues. This oxidized form of NPGPx binds to glucose-regulated protein (GRP)78 and forms covalent bonding intermediates between Cys86 of NPGPx and Cys41/Cys420 of GRP78. Subsequently, the formation of the disulfide bond between Cys41 and Cys420 of GRP78 enhances its chaperone activity. NPGPx-deficient cells display increased reactive oxygen species, accumulated misfolded proteins, and impaired GRP78 chaperone activity. Complete loss of NPGPx in animals causes systemic oxidative stress, increases carcinogenesis, and shortens life span. These results suggest that NPGPx is essential for releasing excessive ER stress by enhancing GRP78 chaperone activity to maintain physiological homeostasis.


Asunto(s)
Proteínas Portadoras/metabolismo , Estrés del Retículo Endoplásmico , Proteínas de Choque Térmico/metabolismo , Estrés Oxidativo , Peroxidasas/metabolismo , Deficiencias en la Proteostasis/enzimología , Transducción de Señal , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cisteína , Daño del ADN , Disulfuros/metabolismo , Relación Dosis-Respuesta a Droga , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Fibroblastos/enzimología , Fibroblastos/patología , Glutatión Peroxidasa , Proteínas de Choque Térmico/genética , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Mutación , Oxidantes/farmacología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Peroxidasas/genética , Unión Proteica , Pliegue de Proteína , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Transfección
3.
J Biomed Sci ; 26(1): 44, 2019 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-31170980

RESUMEN

BACKGROUND: Our previous report suggested that centrosomal P4.1-associated protein (CPAP) is required for Hepatitis B virus (HBV) encoded non-structure protein X (HBx)-mediated nuclear factor kappa light chain enhancer of activated B cells (NF-κB) activation. CPAP is overexpressed in HBV-associated hepatocellular carcinoma (HCC); however, the interaction between CPAP and HBx in HBV-HCC remains unclear. METHODS: The mRNA expression of CPAP and HBx was analyzed by quantitative-PCR (Q-PCR). NF-κB transcriptional activity and CPAP promoter activity were determined using a reporter assay in Huh7 and Hep3B cells. Immunoprecipitation (IP) and in situ proximal ligation assay (PLA) were performed to detect the interaction between CPAP and HBx. Chromatin-IP was used to detect the association of cAMP response element binding protein (CREB) and HBx with the CPAP promoter. Cell proliferation was measured using cell counting kit CCK-8, Bromodeoxyuridine (5-bromo-2'-deoxyuridine, BrdU) incorporation, and clonogenic assays. The tumorigenic effects of CPAP were determined using xenograft animal models. RESULTS: HBx can transcriptionally up-regulate CPAP via interacting with CREB. Overexpressed CPAP directly interacted with HBx to promote HBx-mediated cell proliferation and migration; SUMO modification of CPAP was involved in interacting with HBx. Knocked-down expression of CPAP decreased the HBx-mediated tumorigenic effects, including cytokines secretion. Interestingly, overexpressed CPAP maintained the HBx protein stability in an NF-κB-dependent manner; and the expression levels of CPAP and HBx were positively correlated with the activation status of NF-κB in HCC. Increased expression of CPAP and CREB mRNAs existed in the high-risk group with a lower survival rate in HBV-HCC. CONCLUSION: The interaction between CPAP and HBx can provide a microenvironment to facilitate HCC development via enhancing NF-κB activation, inflammatory cytokine production, and cancer malignancies. This study not only sheds light on the role of CPAP in HBV-associated HCC, but also provides CPAP as a potential target for blocking the hyper-activated NF-κB in HCC.


Asunto(s)
Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteínas Asociadas a Microtúbulos/farmacología , Transactivadores/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Reguladoras y Accesorias Virales
4.
J Cell Sci ; 127(Pt 17): 3735-44, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24994936

RESUMEN

Epithelial-mesenchymal transition (EMT) is a developmental program that is associated with esophageal squamous cell carcinoma (ESCC) progression and metastasis. Recently, C/EBPß has been reported to be an EMT inducer in cancer. However, the detailed molecular mechanisms remain unclear. Here, we report for the first time, that the truncated CCAAT-enhancer-binding protein ß (C/EBPß) LIP isoform is abnormally overexpressed and correlated with cancer metastasis in clinical specimens of human ESCC. Furthermore, we demonstrate that C/EBPß LIP mediates epithelial growth factor (EGF)-induced EMT and increases migration and invasion of esophageal cancer cells in a manner that is dependent on miR-203 inactivation. Finally, we identified miR-203 as a direct target of C/EBPß LIP. Disruption of C/EBPß LIP attenuated the EGF-mediated decrease in miR-203, whereas overexpression of C/EBPß LIP alone markedly suppressed miR-203. In addition, we demonstrated that C/EBPß LIP inhibited miR-203 transcription by directly interacting with a conserved distal regulatory element upstream of the miR-203 locus, and in doing so, orchestrated chromatin remodeling. In conclusion, our results have revealed a new regulatory mechanism that involves C/EBPß-LIP-mediated downregulation of miR-203, which plays a key role in EMT and metastasis.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Carcinoma de Células Escamosas/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Transición Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/metabolismo , Línea Celular Tumoral , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Carcinoma de Células Escamosas de Esófago , Humanos
5.
Biochem Biophys Res Commun ; 478(2): 873-80, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27530925

RESUMEN

The nucleolus is the cellular site of ribosomal (r)DNA transcription and ribosome biogenesis. The 58-kDa microspherule protein (MSP58) is a nucleolar protein involved in rDNA transcription and cell proliferation. However, regulation of MSP58-mediated rDNA transcription remains unknown. Using a yeast two-hybrid system with MSP58 as bait, we isolated complementary (c)DNA encoding Rad50-interacting protein 1 (RINT-1), as a MSP58-binding protein. RINT-1 was implicated in the cell cycle checkpoint, membrane trafficking, Golgi apparatus and centrosome dynamic integrity, and telomere length control. Both in vitro and in vivo interaction assays showed that MSP58 directly interacts with RINT-1. Interestingly, microscopic studies revealed the co-localization of MSP58, RINT-1, and the upstream binding factor (UBF), a rRNA transcription factor, in the nucleolus. We showed that ectopic expression of MSP58 or RINT-1 resulted in decreased rRNA expression and rDNA promoter activity, whereas knockdown of MSP58 or RINT-1 by siRNA exerted the opposite effect. Coexpression of MSP58 and RINT-1 robustly decreased rRNA synthesis compared to overexpression of either protein alone, whereas depletion of RINT-1 from MSP58-transfected cells enhanced rRNA synthesis. We also found that MSP58, RINT-1, and the UBF were associated with the rDNA promoter using a chromatin immunoprecipitation assay. Because aberrant ribosome biogenesis contributes to neoplastic transformation, our results revealed a novel protein complex involved in the regulation of rRNA gene expression, suggesting a role for MSP58 and RINT-1 in cancer development.


Asunto(s)
Proteínas de Ciclo Celular/genética , ADN Ribosómico/genética , Fibroblastos/metabolismo , Proteínas Nucleares/genética , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , ARN Ribosómico/genética , Proteínas de Unión al ARN/genética , Transcripción Genética , Proteínas de Ciclo Celular/metabolismo , Fraccionamiento Celular , Línea Celular Tumoral , Nucléolo Celular/metabolismo , Citosol/metabolismo , ADN Ribosómico/metabolismo , Fibroblastos/citología , Regulación de la Expresión Génica , Humanos , Proteínas Nucleares/metabolismo , Biogénesis de Organelos , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN Ribosómico/biosíntesis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Técnicas del Sistema de Dos Híbridos
6.
J Biomed Sci ; 22: 6, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25591788

RESUMEN

CCAAT/enhancer-binding protein delta (CEBPD) belongs to the CCAAT/enhancer-binding protein family, and these proteins function as transcription factors in many biological processes, including cell differentiation, motility, growth arrest, proliferation, cell death, metabolism and immune responses. The functional diversity of CEBPD depends, in part, on the cell type and cellular context, which indicates that CEBPD could interpret a variety of cues to adjust cellular responses in specific situations. Here, we review the regulation of the CEBPD gene and its function in response to inflammatory stimuli. We also address its effects in inflammation-related diseases through a discussion of its recently discovered downstream targets. Regarding to the previous discoveries and new insights in inflammation-associated diseases, suggesting CEBPD could also be a central gene in inflammation. Importantly, the results of this study indicate that the investigation of CEBPD could open a new avenue to help better understand the inflammatory response.


Asunto(s)
Proteína delta de Unión al Potenciador CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Inflamación/genética , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo
7.
Environ Toxicol ; 30(9): 1024-32, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24639330

RESUMEN

Lead ions (Pb(2+) ) are toxic industrial pollutants associated with chronic inflammatory diseases in humans and animals. Previously, we found that Pb(2+) ions induce COX-2 gene expression via the EGF receptor/nuclear factor-κB signal transduction pathway in epidermoid carcinoma cell line A431. In this study, to see whether Pb(2+) ions affect COX-2 expression by epigenetic mechanisms, we looked at the mRNAs of DNA methyltransferases (DNMTs) using real-time PCR of total RNA from these cells. Cells exposed to Pb(2+) had low levels of DNMT3a mRNA, whereas the levels of DNMT1 and DNMT3b mRNAs remained unchanged. Pretreatment of cells with DNMT inhibitor 5-aza-2'-deoxycytidine (5 µM) followed by Pb(2+) (1 µM) significantly increased levels of COX-2 mRNA compared with cells treated with Pb(2+) alone. Overexpression of tumor suppressor gene Rb correlated with an increase in COX-2 mRNA and a decrease in DNMT3a mRNA. Conversely, overexpression of transcription factor E2F1 correlated with a decrease in COX-2 mRNA and an increase in DMNT3a mRNA. Pretreatment with EGFR inhibitors AG1478 and PD153035 significantly limited Pb(2+) -induced reduction in DNMT3a mRNA. In addition, gene knockdown of DNMT3a with short hairpin RNA correlated with increased COX-2 mRNA induced by Pb(2+) . Our findings suggest Pb(2+) ions induce COX-2 expression indirectly by reducing DNMT3a methylation of the COX-2 promoter via transcription factors Rb and E2F1.


Asunto(s)
Ciclooxigenasa 2/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Contaminantes Ambientales/toxicidad , Plomo/toxicidad , Transcriptoma/efectos de los fármacos , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Ciclooxigenasa 2/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/efectos de los fármacos , ADN Metiltransferasa 3A , Decitabina , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Humanos , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , ADN Metiltransferasa 3B
8.
EMBO J ; 29(24): 4106-17, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21076392

RESUMEN

Inflammation and hypoxia are known to promote the metastatic progression of tumours. The CCAAT/enhancer-binding protein-δ (C/EBPδ, CEBPD) is an inflammatory response gene and candidate tumour suppressor, but its physiological role in tumourigenesis in vivo is unknown. Here, we demonstrate a tumour suppressor function of C/EBPδ using transgenic mice overexpressing the Neu/Her2/ERBB2 proto-oncogene in the mammary gland. Unexpectedly, this study also revealed that C/EBPδ is necessary for efficient tumour metastasis. We show that C/EBPδ is induced by hypoxia in tumours in vivo and in breast tumour cells in vitro, and that C/EBPδ-deficient cells exhibit reduced glycolytic metabolism and cell viability under hypoxia. C/EBPδ supports CXCR4 expression. On the other hand, C/EBPδ directly inhibits expression of the tumour suppressor F-box and WD repeat-domain containing 7 gene (FBXW7, FBW7, AGO, Cdc4), encoding an F-box protein that promotes degradation of the mammalian target of rapamycin (mTOR). Consequently, C/EBPδ enhances mTOR/AKT/S6K1 signalling and augments translation and activity of hypoxia-inducible factor-1α (HIF-1α), which is necessary for hypoxia adaptation. This work provides new insight into the mechanisms by which metastasis-promoting signals are induced specifically under hypoxia.


Asunto(s)
Proteína delta de Unión al Potenciador CCAAT/metabolismo , Proteínas F-Box/biosíntesis , Regulación de la Expresión Génica , Hipoxia , Neoplasias Mamarias Animales/secundario , Metástasis de la Neoplasia/patología , Ubiquitina-Proteína Ligasas/biosíntesis , Animales , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Proteína 7 que Contiene Repeticiones F-Box-WD , Glucólisis , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/fisiopatología , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia/fisiopatología
9.
Biochem Biophys Res Commun ; 446(1): 267-71, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24589738

RESUMEN

Recent clinical study indicated that up-regulation of miR-146b was associated with poor overall survival of patients in esophageal squamous cell carcinoma. However, the underlying mechanism of miR-146b dysregulation remains to be explored. Here we report that miR-146b promotes cell proliferation and inhibits cell apoptosis in esophageal cancer cell lines. Mechanismly, two C/EBPß binding motifs are located in the miR-146b promoter conserved region. Among the three isoforms of C/EBPß, C/EBPß LAP2 positively regulated miR-146b expression and increases miR-146b levels in a dose-dependent manner through transcription activation of miR-146b gene. Together, these results suggest a miR-146b regulatory mechanism involving C/EBPß, which may contribute to the up-regulation of miR-146b in esophageal squamous cell carcinoma.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Apoptosis , Secuencia de Bases , Sitios de Unión/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Secuencia Conservada , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Regulación Neoplásica de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Isoformas de Proteínas/metabolismo , Activación Transcripcional , Regulación hacia Arriba
10.
Cancer Cell ; 10(1): 13-24, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843262

RESUMEN

BRCA1 exerts transcriptional repression through interaction with CtIP in the C-terminal BRCT domain and ZBRK1 in the central domain. A dozen genes, including angiopoietin-1 (ANG1), a secreted angiogenic factor, are corepressed by BRCA1 and CtIP based on microarray analysis of mammary epithelial cells in 3D culture. BRCA1, CtIP, and ZBRK1 form a complex that coordinately represses ANG1 expression via a ZBRK1 recognition site in the ANG1 promoter. Impairment of this complex upregulates ANG1, which stabilizes endothelial cells that form a capillary-like network structure. Consistently, Brca1-deficient mouse mammary tumors exhibit accelerated growth, pronounced vascularization, and overexpressed ANG1. These results suggest that, besides its role in maintaining genomic stability, BRCA1 directly regulates the expression of angiogenic factors to modulate the tumor microenvironment.


Asunto(s)
Angiopoyetina 1/genética , Proteína BRCA1/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias Mamarias Experimentales/patología , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Represoras/metabolismo , Animales , Proteína BRCA1/genética , Línea Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Endodesoxirribonucleasas , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Mutación/genética , Neovascularización Patológica/patología , Unión Proteica , Interferencia de ARN , Elementos de Respuesta/genética
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(5): 159492, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38575107

RESUMEN

Obesity is one of the significant health challenges in the world and is highly associated with abnormal adipogenesis. TG-interacting factor 1 (TGIF1) is essential for differentiating murine adipocytes and human adipose tissue-derived stem cells. However, the mode of action needs to be better elucidated. To investigate the roles of TGIF1 in differentiation in-depth, CRISPR/Cas9 knockout technology was performed to generate TGIF1-silenced preadipocytes. The absence of TGIF1 in 3 T3-F442A preadipocytes abolished lipid accumulation throughout the differentiation using Oil Red O staining. Conversely, we established 3 T3-F442A preadipocytes stably expressing TGIF1 and doxycycline-inducible TGIF1 in TGIF1-silenced 3 T3-F442A preadipocytes. Remarkably, the induction of TGIF1 by doxycycline during the initial differentiation phase successfully promoted lipid accumulation in TGIF1-silenced 3 T3-F442A cells. We further explored the mechanisms of TGIF1 in early differentiation. We demonstrated that TGIF1 promoted the mitotic clonal expansion via upregulation of CCAAT/enhancer-binding proteins ß expression, interruption with peroxisome proliferators activated receptor γ downstream regulation, and inhibition of p27kip1 expression. In conclusion, we strengthen the pivotal roles of TGIF1 in early differentiation, which might contribute to resolving obesity-associated metabolic syndromes.


Asunto(s)
Adipocitos , Adipogénesis , Diferenciación Celular , Proteínas de Homeodominio , Proteínas Represoras , Animales , Humanos , Ratones , Adipocitos/metabolismo , Adipocitos/citología , Adipogénesis/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Mitosis/genética , PPAR gamma/metabolismo , PPAR gamma/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
12.
FEBS Lett ; 598(8): 945-955, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38472156

RESUMEN

TG-interacting factor 1 (TGIF1) contributes to the differentiation of murine white preadipocyte and human adipose tissue-derived stem cells; however, its regulation is not well elucidated. Insulin is a component of the adipogenic cocktail that induces ERK signaling. TGIF1 phosphorylation and sustained stability in response to insulin were reduced through the use of specific MEK inhibitor U0126. Mutagenesis at T235 or T239 residue of TGIF1 in preadipocytes led to dephosphorylation of TGIF1. The reduced TGIF1 stability resulted in an increase in p27kip1 expression, a decrease in phosphorylated Rb expression and cellular proliferation, and a reduced accumulation of lipids compared to the TGIF1-overexpressed cells. These findings highlight that insulin/ERK-driven phosphorylation of the T235 or T239 residue at TGIF1 is crucial for adipocyte differentiation.


Asunto(s)
Células 3T3-L1 , Adipocitos , Adipogénesis , Diferenciación Celular , Proteínas de Homeodominio , Insulina , Animales , Ratones , Fosforilación/efectos de los fármacos , Insulina/metabolismo , Adipocitos/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Humanos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proliferación Celular/efectos de los fármacos , Butadienos/farmacología
13.
J Biomed Sci ; 20: 98, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24359566

RESUMEN

BACKGROUND: Zinc finger protein 179 (Znf179), also known as ring finger protein 112 (Rnf112), is a member of the RING finger protein family and plays an important role in neuronal differentiation. To investigate novel mechanisms of Znf179 regulation and function, we performed a yeast two-hybrid screen to identify Znf179-interacting proteins. RESULTS: Using a yeast two-hybrid screen, we have identified promyelocytic leukemia zinc finger (Plzf) as a specific interacting protein of Znf179. Further analysis showed that the region containing the first two zinc fingers of Plzf is critical for its interaction with Znf179. Although the transcriptional regulatory activity of Plzf was not affected by Znf179 in the Gal4-dependent transcription assay system, the cellular localization of Znf179 was changed from cytoplasm to nucleus when Plzf was co-expressed. We also found that Znf179 interacted with Plzf and regulated Plzf protein expression. CONCLUSIONS: Our results showed that Znf179 interacted with Plzf, resulting in its translocation from cytoplasm to the nucleus and increase of Plzf protein abundance. Although the precise nature and role of the Znf179-Plzf interaction remain to be elucidated, both of these two genes are involved in the regulation of neurogenesis. Our finding provides further research direction for studying the molecular functions of Znf179.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Inmunoprecipitación , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Técnicas del Sistema de Dos Híbridos
14.
Mol Neurobiol ; 60(4): 2200-2208, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36633805

RESUMEN

Astroglial-fibrotic scars resulted from spinal cord injury affect motor and sensory function, leading to paralysis. In particular, the fibrotic scar is a main barrier that disrupts neuronal regeneration after spinal cord injury. However, the association between astrocytes and fibrotic scar formation is not yet understood. We have previously demonstrated that the transcriptional factor Cebpd contributes to astrogliosis, which promotes glial scar formation after spinal cord injury. Herein, we show that fibrotic scar formation was decreased in the epicenter region in Cebpd-/- mice after contusive spinal cord injury and astrocytic Cebpd promoted fibroblast migration through secretion of Ptx3. Furthermore, the expression of Mmp3 was increased under recombinant protein Ptx3 treatment in fibroblasts by observing microarray data, resulting in fibroblast migration. In addition, regulation of Mmp3 occurs through the NFκB signaling pathway by using an irreversible inhibitor of IκBα phosphorylation in pretreated fibroblasts. Of note, we used the synthetic peptide RI37, which blocks fibroblast migration and decreases fibroblast Mmp3 expression in IL-1ß-treated astrocyte conditioned media. Collectively, our data suggest that fibroblast migration can be affected by astrocytic Cebpd through the Ptx3/NFκB/Mmp3 axis pathway and that the RI37 peptide may act as a therapeutic medicine to inhibit fibrotic scar formation after spinal cord injury.


Asunto(s)
Cicatriz , Traumatismos de la Médula Espinal , Ratones , Animales , Cicatriz/patología , Astrocitos/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Traumatismos de la Médula Espinal/patología , Fibrosis , Gliosis/patología , Médula Espinal/patología
15.
Biomed Pharmacother ; 157: 113962, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36370523

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) patients suffer varying degrees of heart dysfunction after tyrosine kinase inhibitor (TKI) treatment. Interestingly, HCC patients often have higher levels of pentraxin 3 (PTX3), and PTX3 inhibition was found to improve left ventricular dysfunction in animal models. OBJECTIVES: We sought to assess the therapeutic potential of PTX3 inhibition on TKI-associated cardiotoxicity. METHODS: We used a human embryonic stem cell line, RUES2, to generate cardiomyocyte cultures (RUES2-CM) for functional testing. We also assessed heart function and PTX3 expression levels in 16 HCC patients who received TKI treatment, 3 HCC patients who did not receive TKIs, and 7 healthy volunteers. RESULTS: Significantly higher PTX3 expression was noted in HCC patients with TKI treatment versus those without, and 38% of male and 33% of female patients had QTc prolongation after TKI treatment. Treatment of cardiomyocyte cultures with sorafenib also increased PTX3 expression and induced cytoskeletal remodelling, contraction reduction, sodium current inhibition, and mitochondrial respiratory dysfunction. PTX3 colocalised with CD44 in cardiomyocytes, and cardiomyocyte contraction, mitochondrial respiratory function, and regular cytoskeletal and apoptotic protein expression were restored with PTX3 inhibition. CD44 knockdown confirmed PTX3/CD44 signalling. These results suggest a possible mechanism in which sorafenib treatment increases PTX3 expression, thereby resulting in reduced extracellular signal-regulated kinase (ERK) 1/2 expression that affects cardiomyocyte contraction, while also activating c-Jun N-terminal kinase (JNK) downstream pathways to disrupt mitochondrial respiration and trigger apoptosis. CONCLUSIONS: TKI-induced cardiotoxicity may be partly mediated by the upregulation of PTX3, and thus PTX3 inhibition has potential as a therapeutic strategy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Humanos , Masculino , Femenino , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Proteína C-Reactiva/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Sorafenib/uso terapéutico , Cardiotoxicidad , Mitocondrias/metabolismo
16.
J Biol Chem ; 286(33): 28662-28670, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21715338

RESUMEN

Epidemiologic and clinical research indicates that chronic inflammation increases the risk of certain cancers, possibly through chromosomal instability. However, the mechanism of inflammation-dependent chromosomal instability associated with tumorigenesis is not well characterized. The transcription factor CCAAT/enhancer-binding protein δ (C/EBPδ, CEBPD) is induced by tumor necrosis factor α (TNFα) and expressed in chronically inflamed tissue. In this study, we show that TNFα promotes aneuploidy. Loss of CEBPD attenuated TNFα-induced aneuploidy, and CEBPD caused centromere abnormality. Additionally, TNFα-induced CEBPD expression augmented anchorage-independent growth. We found that TNFα induced expression of aurora kinase C (AURKC) through CEBPD, and that AURKC also causes aneuploidy. Furthermore, high CEBPD expression correlated with AURKC expression in inflamed cervical tissue specimens. These data provide insight into a novel function for CEBPD in inducing genomic instability through the activation of AURKC expression in response to inflammatory signals.


Asunto(s)
Proteína delta de Unión al Potenciador CCAAT/metabolismo , Cuello del Útero/metabolismo , Inestabilidad Genómica , Proteínas Serina-Treonina Quinasas/biosíntesis , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo , Cervicitis Uterina/metabolismo , Aneuploidia , Animales , Aurora Quinasa C , Aurora Quinasas , Proteína delta de Unión al Potenciador CCAAT/genética , Centrómero/genética , Centrómero/metabolismo , Centrómero/patología , Cuello del Útero/patología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Células HeLa , Humanos , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Factor de Necrosis Tumoral alfa/farmacología , Cervicitis Uterina/genética , Cervicitis Uterina/patología
17.
Ann Surg Oncol ; 19(2): 443-54, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21761100

RESUMEN

BACKGROUND: The understanding of epidermal growth factor receptor (EGFR) deregulation in carcinogenesis remains incomplete. We investigated the implications of EGFR gene status and EGFR nuclear translocation in gallbladder carcinoma (GBCA). METHODS: Subcellular localization of EGFR and phosphorylated EGFR (pEGFR) was analyzed by fractional immunoblotting and confocal immunofluorescence in GBCA cell lines. pEGFR binding to iNOS promoter was assessed by chromatin immunoprecipitation with iNOS promoter activity evaluated by luciferase assay. EGFR, pEGFR, and iNOS were immunohistochemically assessable for localization and level in the training set of 104 GBCAs on tissue microarrays, with 76 cases analyzed for EGFR gene by chromogenic in situ hybridization (CISH) and mutant-enriched PCR targeting exons 19 and 21. The prognostic impact of nuclear pEGFR (N-pEGFR) immunoexpression was reaffirmed on whole sections of 58 GBCAs in the test set. RESULTS: Nuclear expression of EGFR and pEGFR was substantiated in vitro with augmented activity of iNOS promoter elicited by pEGFR binding upon EGF treatment. Despite no mutation, EGFR amplification, identified in 11 cases (15%) by CISH, strongly correlated with cytoplasmic EGFR expression (P < 0.001) but not with disease-specific survival (DSS). Immunoexpression of nuclear EGFR (N-EGFR), cytoplasmic pEGFR, and N-pEGFR was strongly related to that of iNOS (all ≤0.005). N-pEGFR independently predicted worse DSS in both training (P = 0.0468, HR = 2.024) and test sets (P = 0.0223, HR = 5.573). CONCLUSIONS: N-EGFR and N-pEGFR express in GBCA, conferring clinical aggressiveness partly through iNOS transactivation. Lacking response-predicting mutation, EGFR gene status, albeit amplified in 15% of GBCA, is neither related to nuclear EGFR translocation nor prognostically useful.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma Papilar/metabolismo , Núcleo Celular/metabolismo , Receptores ErbB/metabolismo , Neoplasias de la Vesícula Biliar/metabolismo , Regulación Neoplásica de la Expresión Génica , Óxido Nítrico Sintasa de Tipo II/genética , Transporte Activo de Núcleo Celular , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Anciano , Western Blotting , Carcinoma Papilar/genética , Carcinoma Papilar/mortalidad , Inmunoprecipitación de Cromatina , Citoplasma/metabolismo , Receptores ErbB/genética , Femenino , Técnica del Anticuerpo Fluorescente , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/mortalidad , Humanos , Técnicas para Inmunoenzimas , Hibridación in Situ , Luciferasas/metabolismo , Masculino , Mutación/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Pronóstico , Regiones Promotoras Genéticas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Tasa de Supervivencia , Células Tumorales Cultivadas , Regulación hacia Arriba
18.
J Pathol ; 225(2): 243-54, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21710690

RESUMEN

Over-expression of AURKC has been detected in human colorectal cancers, thyroid carcinoma and several cancer cell lines. However, the regulation and clinical implications of over-expressed AURKC in cancer cells are unclear. Here we show that elevated AURKC increases the proliferation, transformation and migration of cancer cells. Importantly, the kinase activity of AURKC is required for these tumour-associated properties. Analysis of human cancer specimens shows that the expression of AURKC is increased in cervical cancer, and is highly correlated with staging in colorectal cancer. Over-expressed AURKC-GFP localizes to the centromeric regions of mitotic chromosomes and results in a decreased level of AURKB, a key regulator of spindle checkpoint. Expression of AURKC is down-regulated by PLZF, a transcriptional repressor, through recruitment to its promoter region. The expression levels of PLZF and AURKC mRNA display opposite patterns in human cervical and colorectal cancers. Taken together, our results provide important insights into human cancers with AURKC expression, which may serve as a potential target for cancer therapy in the future.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Serina-Treonina Quinasas/biosíntesis , Animales , Aurora Quinasa B , Aurora Quinasa C , Aurora Quinasas , Movimiento Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Células Epiteliales/patología , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Serina-Treonina Quinasas/genética , Regulación hacia Arriba
19.
J Immunol Res ; 2022: 8953235, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36530573

RESUMEN

Background: Since food avoidance is currently the only way to prevent allergic reactions to shrimp, a better understanding of molecular events in the induction and progression of allergy, including food allergy, is needed for developing strategies to inhibit allergic responses. Pentraxin 3 (PTX3) is rapidly produced directly from inflammatory or damaged tissues and is involved in acute immunoinflammatory responses. However, the role of PTX3 in the development of immediate IgE-mediated shrimp allergy remains unknown. Methods: Wild-type BALB/c mice were immunized intraperitoneally and were challenged with shrimp extract. Serum IgE and PTX3 levels were analyzed. RBL-2H3 cells were stimulated with either dinitrophenyl (DNP) or serum of shrimp-allergic mice, and markers of degranulation, proinflammatory mediators, and phosphorylation of signal proteins were analyzed. We further examined the effect of PTX3 in shrimp extract-induced allergic responses in vitro and in vivo. Results: Mice with shrimp allergy had increased PTX3 levels in the serum and small intestine compared with healthy mice. PTX3 augmented degranulation, the production of proinflammatory mediators, and activation of the Akt and MAPK signaling pathways in mast cells upon DNP stimulation. Furthermore, the expression of transcription factor CCAAT/enhancer-binding protein delta (CEBPD) was elevated in PTX3-mediated mast cell activation. Finally, the PTX3 inhibitor RI37 could attenuate PTX3-induced degranulation, proinflammatory mediator expression, and phosphorylation of the Akt and MAPK signaling. Conclusions: The results suggested that PTX3 can facilitate allergic responses. Our data provide new insight to demonstrate that PTX3 is a cause of allergic inflammation and that RI37 can serve as a therapeutic agent in shrimp allergy.


Asunto(s)
Hipersensibilidad , Mastocitos , Ratones , Animales , Inmunoglobulina E , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones Endogámicos BALB C , Degranulación de la Célula
20.
Anal Chim Acta ; 1203: 339722, 2022 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-35361435

RESUMEN

An integrated microfluidic Au nanoparticle (AuNP) aptasensor device is proposed for monitoring the concentration of potassium (K+) ions in the bloodstream of patients with chronic kidney disease (CKD). In the proposed detection device, the AuNPs in the AuNP/aptamer complex are displaced by the serum K+ ions and react with NaCl to produce a color change in the detection region from which the K+ ion concentration is then inversely derived. The microfluidic device comprises two main components, namely an AuNP aptasensor PMMA (Poly(methyl methacrylate))/paper-microchip and a colorimetric analysis system for the quantitative detection of K+ ion concentration in whole blood. The functions of PMMA/paper microchips include reagent storage, K+ ion/aptamer reaction, and separation of serum from whole blood samples (blood filter). Experimental results show that the microfluidic device provides a linear response over the K+ ion concentration in range of 0.05-9 mM in artificial serum and has a detection limit (LOD) of 0.01 mM. Moreover, the detection results obtained for the 137 whole blood and 287 serum samples of CKD patients are very consistent (R2 = 0.968 and R2 = 0.980) with the measurement results obtained using an ion-selective electrodes (ISE) method. Results confirm that the current microfluidic aptasensor device provides a highly-sensitive and convenient method for performing the point-of-care (POC) monitoring of the whole blood K+ ion concentration.


Asunto(s)
Aptámeros de Nucleótidos , Técnicas Biosensibles , Nanopartículas del Metal , Técnicas Biosensibles/métodos , Oro , Humanos , Iones , Dispositivos Laboratorio en un Chip , Microfluídica , Sistemas de Atención de Punto , Potasio/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA