Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Genes Dev ; 30(18): 2093-2105, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798850

RESUMEN

Glucocorticoid (GC) receptor (GR) has been shown recently to bind a subset of mRNAs and elicit rapid mRNA degradation. However, the molecular details of GR-mediated mRNA decay (GMD) remain unclear. Here, we demonstrate that GMD triggers rapid degradation of target mRNAs in a translation-independent and exon junction complex-independent manner, confirming that GMD is mechanistically distinct from nonsense-mediated mRNA decay (NMD). Efficient GMD requires PNRC2 (proline-rich nuclear receptor coregulatory protein 2) binding, helicase ability, and ATM-mediated phosphorylation of UPF1 (upstream frameshift 1). We also identify two GMD-specific factors: an RNA-binding protein, YBX1 (Y-box-binding protein 1), and an endoribonuclease, HRSP12 (heat-responsive protein 12). In particular, using HRSP12 variants, which are known to disrupt trimerization of HRSP12, we show that HRSP12 plays an essential role in the formation of a functionally active GMD complex. Moreover, we determine the hierarchical recruitment of GMD factors to target mRNAs. Finally, our genome-wide analysis shows that GMD targets a variety of transcripts, implicating roles in a wide range of cellular processes, including immune responses.


Asunto(s)
Monocitos/metabolismo , Estabilidad del ARN/fisiología , Receptores de Glucocorticoides/metabolismo , Adenosina Trifosfatasas/metabolismo , Quimiocina CCL2/metabolismo , Quimiotaxis/genética , Células HEK293 , Células HeLa , Proteínas de Choque Térmico/metabolismo , Humanos , Monocitos/enzimología , Monocitos/inmunología , Fosforilación , Polimerizacion , ARN Helicasas , Estabilidad del ARN/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Ribonucleasas/metabolismo , Transactivadores/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo
2.
Carcinogenesis ; 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38085813

RESUMEN

Tumor-associated macrophages (TAMs) are among the most abundant cell types in the tumor microenvironment (TME). The immunosuppressive TME formed by TAMs is an essential prerequisite for cancer progression. Tumor-derived microvesicles (MVs), a subtype of extracellular vesicle shed directly from the plasma membrane, are important regulators of intercellular communication and TME modulation during tumorigenesis. However, the exact mechanism by which tumor-derived MVs induce the generation of the immunosuppressive TME and polarization of TAMs remains unclear. Here, we investigated the role of CD133-containing MVs derived from colorectal cancer (CRC) cells in macrophage polarization and cancer progression. CD133-containing MVs from CRC cells were incorporated into macrophages, and M0 macrophages were morphologically transformed into M2-like TAMs. CD133-containing MVs were found to increase the mRNA expression of M2 macrophage markers. Additionally, cytokine array analysis revealed that M2-like TAMs induced by CD133-containing MVs increased the secretion of interleukin 6, which activated the STAT3 pathway in CRC cells. Furthermore, the conditioned medium of M2-like TAMs promoted cell motility, epithelial-mesenchymal transition, and cell proliferation. However, MVs from CD133-knockdown cells had little effect on TAM polarization and CRC progression. These results demonstrate that CD133-containing MVs induce M2-like TAM polarization and contribute to cancer progression by mediating crosstalk between tumor cells and TAMs in the TME of CRC.

3.
Cell Mol Life Sci ; 79(9): 505, 2022 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-36057892

RESUMEN

In multiple cancers, autophagy promotes tumor development by recycling intracellular components into metabolic pathways. Autophagy-induced metabolic reprogramming and plasticity lead to cancer cell survival and resistance to anticancer therapy. We investigated the role of small leucine zipper protein (sLZIP) in autophagy and cell survival under nutrient-deficient conditions in colorectal cancer (CRC). sLZIP was induced by nutrient stress and increased the transcription of microtubule-associated protein 1A/1B-light chain 3 (LC3), by directly binding to its promoter. Under nutrient stress conditions, sLZIP activated autophagy and promoted the survival of CRC cells. sLZIP induced metabolic reprogramming of CRC cells, to activate glutaminolysis and the tricarboxylic acid cycle. sLZIP also enhanced the autophagic degradation of Keap1 and the nuclear accumulation of Nrf2, leading to NQO1 expression, for maintenance of redox homeostasis. sLZIP-knockout CRC cells exhibited impaired autophagy induction in the glycolytic inhibition state. Xenograft mice lacking sLZIP showed decreased tumor growth, by rendering CRC cells sensitive to glycolysis inhibition. The expression of sLZIP and LC3B was highly elevated in tumors of CRC patients compared to that in normal tissues, and correlated with the progression of CRC. These findings suggest that sLZIP drives autophagy and metabolic reprogramming to promote colorectal tumorigenesis.


Asunto(s)
Neoplasias Colorrectales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Animales , Autofagia , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Leucina Zippers , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Nutrientes
4.
J Cell Physiol ; 237(12): 4504-4516, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36250997

RESUMEN

Chronic liver injury follows inflammation and liver fibrosis; however, the molecular mechanism underlying fibrosis has not been fully elucidated. In this study, the role of ductal WW domain-containing transcription regulator 1 (WWTR1)/transcriptional coactivator with PDZ-binding motif (TAZ) was investigated after liver injury. Ductal TAZ-knockout (DKO) mice showed decreased liver fibrosis following a Diethyl 1,4-dihydro-2,4,6-trimethyl-3,5-pyridinedicarboxylate (DDC) diet compared to wild-type (WT) mice, as evidenced by decreased expression levels of fibrosis inducers, including connective tissue growth factor (Ctgf)/cellular communication network factor 2 (CCN2), cysteine-rich angiogenic inducer 61 (Cyr61/CCN1), and transforming growth factor beta 1 (Tgfb1), in DKO mice. Similarly, TAZ-knockout (KO) cholangiocyte organoids showed decreased expression of fibrosis inducers. Additionally, the culture supernatant of TAZ-KO cholangiocyte organoids decreased the fibrogenic gene expression in liver stellate cells. Further studies revealed that prominin 1 (PROM1/CD133) stimulated TAZ for fibrosis. After the administration of DDC diet, fibrosis was decreased in CD133-KO (CD133-KO) mice compared to that in WT mice. Similarly, CD133-KO cholangiocyte organoids showed decreased Ctgf, Cyr61, and Tgfb1 expression levels compared to WT cholangiocyte organoids. Mechanistically, CD133 stabilized TAZ via Src activation. Inhibition of Src decreased TAZ levels. Similarly, CD133-knockdown HCT116 cells showed decreased TAZ levels, but reintroduction of active Src recovered the TAZ levels. Taken together, our results suggest that TAZ facilitates liver fibrosis after a DDC diet via the CD133-Src-TAZ axis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Transactivadores , Animales , Ratones , Dieta , Fibrosis , Péptidos y Proteínas de Señalización Intracelular , Hígado , Cirrosis Hepática/inducido químicamente , Ratones Noqueados , Factores de Transcripción/genética , Proteínas Proto-Oncogénicas pp60(c-src) , Proteínas Adaptadoras Transductoras de Señales/genética
5.
Carcinogenesis ; 42(8): 1089-1099, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34050742

RESUMEN

Matrix metalloproteinases (MMPs) function as central modulators of tissue remodeling. Abnormal expression and altered activity of MMPs result in excessive extracellular matrix degradation and increased tumor metastasis in various cancers. Small leucine zipper protein (sLZIP), belonging to the leucine zipper transcription factor family, functions as a transcriptional regulator of genes involved in various cellular processes. However, its role in MMP expression and castration-resistant prostate cancer (CRPC) metastasis remains unclear. In this study, we investigated the role of sLZIP in MMP-13 expression and its involvement in CRPC metastasis. sLZIP increased MMP-13 transcription by directly binding to its promoter in CRPC cells. We found that the expression levels of glucocorticoid receptor (GR), which represses MMP transcription, were elevated in CRPC cells. However, sLZIP suppressed the inhibitory effect of GR and enhanced the secretion of MMP-13 in CRPC cells. sLZIP promoted cell migration and invasion; however, a specific MMP-13 inhibitor blocked sLZIP-induced cell motility. Depletion of sLZIP using the CRISPR/Cas9 system downregulated MMP-13 messenger RNA expression in PC3 cells. Mice injected with sLZIP-depleted PC3 cells showed significantly reduced metastatic tumor volume in the lung compared with mice injected with control PC3 cells. Our findings suggest that sLZIP plays an important role in MMP-13 induction and CRPC metastasis. Therefore, sLZIP inhibition could be a novel therapeutic strategy for metastatic GR-enriched CRPC.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Metaloproteinasa 13 de la Matriz/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Transcripción Genética/fisiología , Animales , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Regiones Promotoras Genéticas
6.
Carcinogenesis ; 41(7): 940-949, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-31584624

RESUMEN

Cancer stem cells (CSCs) initiate tumors and possess the properties of self-renewal and differentiation. Since they are responsible for chemoresistance, CSCs are known to be a key factor in cancer recurrence. α-Actinin-4 (ACTN4) is an actin-binding protein that is involved in muscle differentiation and cancer metastasis. It promotes epithelial to mesenchymal transition and cell cycle progression via ß-catenin stabilization in cervical cancer. In the present study, we investigated the role of ACTN4 in regulating cancer cell stemness and chemoresistance in cervical cancer. Results from the gene expression database analysis showed that ACTN4 mRNA expression was elevated in cancerous cervices when compared with normal cervices. Furthermore, ACTN4 knockdown suppressed sphere formation and CSC proliferation. It also decreased CSC size and CD44high/CD24low cell population. ACTN4-knockdown CSCs were sensitive to anticancer drugs, which was observed by down-regulation of the ATP-binding cassette family G2 involved in drug resistance. Finally, ACTN4-knockdown CSCs formed reduced tumors in vivo when compared with control CSCs. Overall, these findings suggest that ACTN4 regulates CSC properties and contributes to chemoresistance in cervical cancer.


Asunto(s)
Actinina/genética , Antineoplásicos/farmacología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Antineoplásicos/efectos adversos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Ratones , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Esferoides Celulares/efectos de los fármacos , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
FASEB J ; 33(3): 4248-4260, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30521383

RESUMEN

Extracellular vesicles contain various cellular components that are involved in tumor growth, metastasis, and immune escape. Extracellular vesicles are classified into 2 groups, namely, exosomes and microvesicles (MV). Although the formation and roles of exosomes have been studied, the exact functions of MVs and mechanisms underlying MV release are not fully understood. We found that epidermal growth factor accelerates the release of MVs from the plasma membrane by inducing NF-κB activation and CD133 expression. The amount and sizes of budding MVs were found to be dependent on the expression level of CD133, which regulates the activities of the small guanosine 5'-triphosphatases RhoA and Rac1. CD133-containing MVs released from KRAS mutant colon cancer cells delivered KRAS mutant to adjacent nontumorigenic cells and activated KRAS downstream signaling. CD133-containing MVs were found to promote the migration and invasion of adjacent cells. CD133-containing MVs induced the development of chemoresistance by abolishing the inhibitory effects of anti-epidermal growth factor receptor (EGFR) drugs on cell proliferation and motility in colon cancer. These results suggest that CD133 acts as a novel modulator in MV release and in oncoprotein trafficking. CD133 can serve as a therapeutic target for treatment of anti-EGFR drug-resistant colon cancer.-Kang, M., Kim, S., Ko, J. Roles of CD133 in microvesicle formation and oncoprotein trafficking in colon cancer.


Asunto(s)
Antígeno AC133/metabolismo , Neoplasias del Colon/metabolismo , Proteínas Oncogénicas/metabolismo , Animales , Membrana Celular/metabolismo , Membrana Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/fisiología , Receptores ErbB/metabolismo , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/fisiología , Células HCT116 , Células HEK293 , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Transporte de Proteínas/fisiología , Transducción de Señal/fisiología
8.
Biochem Biophys Res Commun ; 501(3): 800-806, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29772231

RESUMEN

The glucocorticoid receptor (GR) is a ligand-dependent transcription factor that mediates the effects of glucocorticoids, and plays a crucial role in cell growth, development, inflammation, and gluconeogenesis. The 14-3-3 proteins bind to target proteins via phosphorylation, and influence many cellular events by altering their subcellular localization or by acting as chaperones. However, the mechanisms by which 14-3-3 proteins regulate GR transactivation and their involvement in gluconeogenesis remain uncharacterized. We found that 14-3-3ß and γ increased GR transcriptional activity and the promoter activities of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase in the presence of glucocorticoids. Inhibition of the endogenous 14-3-3ß and γ decreased dexamethasone- and cAMP-stimulated PEPCK expression. Further, both 14-3-3ß and γ increased glucose production in response to glucocorticoids. Our findings suggest that 14-3-3ß and γ function as positive regulators of GR transactivation and glucocorticoid-mediated hepatic gluconeogenesis.


Asunto(s)
Proteínas 14-3-3/metabolismo , Gluconeogénesis , Hígado/metabolismo , Receptores de Glucocorticoides/genética , Activación Transcripcional , Glucosa/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo
9.
FASEB J ; 31(6): 2548-2561, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28246167

RESUMEN

The molecular mechanism of stress-induced hepatic steatosis is not well known. Human leucine zipper protein (LZIP) regulates the expression of genes involved in inflammation, cell migration, and stress response. The aim of this study was to determine the regulatory role of LZIP in stress-induced hepatic steatosis. We used a microarray analysis to identify LZIP-induced genes involved in hepatic lipid metabolism. LZIP increased the expression of apolipoprotein A-IV (APOA4) mRNA. In the presence of stress inducer, APOA4 promoter analysis was performed, and LZIP-induced lipid accumulation was monitored in mouse primary cells and human tissues. Under Golgi stress conditions, LZIP underwent proteolytic cleavage and was phosphorylated by AKT to protect against proteasome degradation. The stabilized N-terminal LZIP was translocated to the nucleus, where it directly bound to the APOA4 promoter, leading to APOA4 induction. LZIP-induced APOA4 expression resulted in increased absorption of surrounding free fatty acids. LZIP also promoted hepatic steatosis in mouse liver. Both LZIP and APOA4 were highly expressed in human steatosis samples. Our findings indicate that LZIP is a novel modulator of APOA4 expression and hepatic lipid metabolism. LZIP might be a therapeutic target for developing treatment strategies for hepatic steatosis and related metabolic diseases.-Kang, M., Kim, J., An, H.-T., Ko, J. Human leucine zipper protein promotes hepatic steatosis via induction of apolipoprotein A-IV.


Asunto(s)
Apolipoproteínas A/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hígado Graso/metabolismo , Apolipoproteínas A/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Hígado Graso/genética , Regulación de la Expresión Génica/fisiología , Aparato de Golgi/fisiología , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Mutación , Ácido Oléico/metabolismo , Fosforilación , Plásmidos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estrés Fisiológico
10.
Proc Natl Acad Sci U S A ; 112(13): E1540-9, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25775514

RESUMEN

Glucocorticoid receptor (GR), which was originally known to function as a nuclear receptor, plays a role in rapid mRNA degradation by acting as an RNA-binding protein. The mechanism by which this process occurs remains unknown. Here, we demonstrate that GR, preloaded onto the 5'UTR of a target mRNA, recruits UPF1 through proline-rich nuclear receptor coregulatory protein 2 (PNRC2) in a ligand-dependent manner, so as to elicit rapid mRNA degradation. We call this process GR-mediated mRNA decay (GMD). Although GMD, nonsense-mediated mRNA decay (NMD), and staufen-mediated mRNA decay (SMD) share upstream frameshift 1 (UPF1) and PNRC2, we find that GMD is mechanistically distinct from NMD and SMD. We also identify de novo cellular GMD substrates using microarray analysis. Intriguingly, GMD functions in the chemotaxis of human monocytes by targeting chemokine (C-C motif) ligand 2 (CCL2) mRNA. Thus, our data provide molecular evidence of a posttranscriptional role of the well-studied nuclear hormone receptor, GR, which is traditionally considered a transcription factor.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Glucocorticoides/metabolismo , Transactivadores/metabolismo , Quimiocina CCL2/metabolismo , Quimiotaxis , Genes Reporteros , Células HEK293 , Células HeLa , Humanos , Ligandos , Monocitos/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido , Unión Proteica , ARN Helicasas , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo
11.
Biochim Biophys Acta ; 1849(10): 1237-47, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26260846

RESUMEN

Peroxisome proliferator activated receptor (PPAR) γ2 plays important roles in glucose and lipid metabolism in hepatocytes. PPARγ2 is involved in metabolic disorders, including obesity, diabetes, and fatty liver disease. Although the 14-3-3 proteins participate in a variety of cell signal pathways, the roles of the 14-3-3 proteins in regulating PPARγ2 transactivation and hepatic lipid metabolism are unknown. We identified 14-3-3ß and γ as PPARγ2 transcriptional regulators. We found that 14-3-3ß and γ competitively interacted with the phosphorylated Ser273 of PPARγ2, which is important for regulating glucose and lipid metabolism. 14-3-3ß increased the transcriptional activity of PPARγ2 and enhanced the expression levels of PPARγ2 target genes involved in lipogenesis and lipid transport. In contrast, 14-3-3γ decreased PPARγ2 transactivation and reduced the expression levels of PPARγ2 target genes. A high concentration of free fatty acids increased PPARγ2 expression and lipid accumulation. 14-3-3ß enhanced hepatic lipogenesis, which is a major symptom of non-alcoholic fatty liver disease. However, 14-3-3γ suppressed hepatic lipid accumulation in the presence of high free fatty acids. These findings indicate that 14-3-3ß and γ are novel PPARγ2 regulators and are involved in hepatic lipid metabolism. 14-3-3ß and γ can be therapeutic target molecules to treat non-alcoholic fatty liver disease.


Asunto(s)
Proteínas 14-3-3/genética , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , PPAR gamma/metabolismo , Proteínas 14-3-3/metabolismo , Animales , Ácidos Grasos no Esterificados , Glucosa/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Lipogénesis/genética , Hígado/patología , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , PPAR gamma/biosíntesis , PPAR gamma/genética , Cultivo Primario de Células , Transducción de Señal , Activación Transcripcional/genética
12.
J Biol Chem ; 289(8): 4969-79, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24375477

RESUMEN

The small leucine zipper protein (sLZIP) plays a role in transcriptional regulation in various types of cells. However, the role of sLZIP in myogenesis is unknown. We identified α-actinin-4 (ACTN4) as a sLZIP-binding protein. ACTN4 functions as a transcriptional regulator of myocyte enhancer factor (MEF)2, which plays a critical role in expression of muscle-specific genes during skeletal muscle differentiation. We found that ACTN4 translocates to the nucleus, induces myogenic gene expression, and promotes myotube formation during myogenesis. The myogenic process is controlled by an association between myogenic factors and MEF2 transcription factors. ACTN4 increased expression of muscle-specific proteins via interaction with MEF2. However, sLZIP decreased myogenic gene expression and myotube formation during myogenesis via disruption of the association between ACTN4 and MEF2. ACTN4 increased the promoter activities of myogenic genes, whereas sLZIP abrogated the effect of ACTN4 on transcriptional activation of myogenic genes in myoblasts. The C terminus of sLZIP is required for interaction with the C terminus of ACTN4, based on deletion mutant analysis, and sLZIP plays a role in regulation of MEF2 transactivation via interaction with ACTN4. Our results indicate that sLZIP negatively regulates skeletal muscle differentiation via interaction with ACTN4 and that sLZIP can be used as a therapeutic target molecule for treatment of muscle hypertrophy and associated diseases.


Asunto(s)
Actinina/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Diferenciación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Músculo Esquelético/citología , Actinina/química , Secuencia de Aminoácidos , Animales , Línea Celular , Humanos , Factores de Transcripción MEF2/metabolismo , Ratones , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/metabolismo , Especificidad de Órganos , Unión Proteica , Relación Estructura-Actividad , Activación Transcripcional
13.
FASEB J ; 28(11): 5010-21, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25077563

RESUMEN

Atherosclerosis is a chronic inflammatory response of the vascular wall, and immune responses are involved in every phase of atherosclerosis, from initiation, to progression, and finally to plaque rupture. Cytokines are the major atherogenic mediators that promote plaque formation and progression by activation of inflammatory cells. They induce expressions of matrix metalloproteinases (MMPs), leading to vascular smooth muscle cell (VSMC) migration in atherosclerotic lesions. Although chronic inflammatory mediators, including tumor necrosis factor α (TNF-α) and MMPs, exacerbate atherosclerosis, the molecular mechanism of atherogenesis remains unclear. In this study we investigated the role of a novel transcription factor the human small leucine zipper protein (sLZIP) in TNF-α-induced MMP expression, VSMC migration, and atherosclerosis progression. The proinflammatory cytokine TNF-α enhanced sLZIP expression by 3-fold via activation of NF-κB signaling. sLZIP induced MMP-9 transcription and the proteolytic activity of MMP-9 by 2.8- and 3.2-fold (P< 0.05), respectively, in macrophages, leading to enhancement of VSMC migration by 2.7-fold (P<0.005). sLZIP(OE/+) (sLZIP transgenic); LDLR(-/-) mice fed a high-cholesterol diet exhibited enhanced arterial plaque formation and increased VSMC migration from the media into the intima by 2.8- and 2.6-fold (P<0.01), respectively, compared with atherosclerosis-prone LDLR(-/-) mice. These results indicate that human sLZIP plays a critical role in development of atherosclerosis and can be used as a therapeutic target molecule for treatment of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Movimiento Celular/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Células Cultivadas , Regulación Enzimológica de la Expresión Génica , Humanos , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Ratones , Músculo Liso Vascular/citología , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Mol Cell Cardiol ; 72: 157-67, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24650873

RESUMEN

The molecular chaperone heat shock protein 90 (HSP90) is overexpressed in plaques of atherosclerosis patients, and is associated with plaque instability. However, the role of HSP90 in atherosclerosis remains unclear. The present study investigated the effects of HSP90 inhibition on migration and proliferation of vascular smooth muscle cells (VSMCs) and involvement in atherosclerosis. To examine the role of HSP90 in VSMC migration, VSMCs were treated with the specific HSP90 inhibitors, 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) and STA-9090. Results of a chemotaxis assay showed that the HSP90 inhibitors suppress migration of VSMCs. HSP90 inhibition also prevented invasion and sprout formation of VSMCs via inhibition of matrix metalloproteinase-2 proteolytic activity. Results of a flow cytometric analysis showed that HSP90 inhibition induces cell cycle arrest via regulation of cyclin D3, PCNA and pRb. To investigate the role of HSP90 in the development of atherosclerosis, low-density lipoprotein receptor (LDLR) deficient mice were fed with a high cholesterol diet for 4weeks and treated with 17-AAG for 8weeks. HSP90 inhibition suppressed migration of VSMCs into atherosclerotic plaque lesions in high cholesterol diet-stimulated LDLR(-/-) mice. Inhibition of HSP90 attenuates formation of atherosclerotic plaques via suppression of VSMC migration and proliferation, indicating that HSP90 inhibitors can be used as therapeutic agents for atherosclerosis and in stent restenosis.


Asunto(s)
Aterosclerosis/genética , Benzoquinonas/farmacología , Proteínas HSP90 de Choque Térmico/genética , Lactamas Macrocíclicas/farmacología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica/genética , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/metabolismo , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/etiología , Aterosclerosis/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colesterol/administración & dosificación , Colesterol/efectos adversos , Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/etiología , Placa Aterosclerótica/patología , Cultivo Primario de Células , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal , Triazoles/farmacología
15.
Cell Physiol Biochem ; 33(1): 151-64, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24481121

RESUMEN

BACKGROUND/AIMS: Matrix metalloproteinase-9 (MMP-9) plays an important role in tumor invasion and metastasis through the breakdown of extracellular matrix. The c-Jun protein, a major component of the AP-1 transcription factor, is elevated in various cancers. Small leucine zipper protein (sLZIP) is a member of the leucine zipper transcription factor family. Although sLZIP is known to be involved in cancer cell migration and invasion, its biological roles in cancer development and the cellular target genes are not fully understood. In this study, we investigated the role of sLZIP in c-Jun expression, and their effects on expression of MMP-9 and migration of cervical cancer cells. METHODS AND RESULTS: sLZIP up-regulates transcription of c-Jun by binding directly to the CRE region in the c-Jun promoter. Elevated c-Jun due to sLZIP leads to activation of MMP-9 transcription by interaction with the AP-1 binding site in the MMP-9 promoter. c-Jun siRNA repressed migration and invasion of cervical cancer cells, whereas sLZIP recovered migration and invasion of cells transfected with c-Jun siRNA. Immunohistochemical analysis results revealed a significant correlation between the expressions of sLZIP and MMP-9 in clinical cervical specimens. CONCLUSION: These results indicate that sLZIP plays a role in expression of c-Jun, and migration and invasion of cervical cancer cells via regulation of MMP-9 transcription.


Asunto(s)
Movimiento Celular/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-jun/genética , Transcripción Genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Secuencia de Bases , Línea Celular Tumoral , Femenino , Humanos , Metaloproteinasa 9 de la Matriz/genética , Datos de Secuencia Molecular , Invasividad Neoplásica , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Neoplasias del Cuello Uterino/enzimología
16.
Heliyon ; 10(7): e29292, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38601650

RESUMEN

Angiogenesis is an indispensable mechanism in cancer progression, as cancer cells need to establish blood vessels to supply oxygen and nutrients. Extracellular vesicles (EVs) derived from cancer cells act as messengers in the tumor microenvironment and induce resistance to anti-angiogenic cancer treatment. EVs can be classified into two categories: exosomes and microvesicles (MVs). Although exosomes are involved in angiogenesis, the role of MVs in angiogenesis and cancer progression remains unclear. CD133 plays a key role in MV formation and oncoprotein trafficking. In this study, we investigated the role of CD133-containing MVs derived from colorectal cancer (CRC) in angiogenesis and cancer progression. CRC-derived MVs were incorporated into endothelial cells and increased the mesh area and tube length of endothelial cells. CD133-containing MVs also stimulate vessel sprouting in endothelial cell spheroids and mouse thoracic aortas. However, MVs derived from CD133-knockdown CRC cells exerted a limited effect on tube formation and vessel sprouting. CD133-containing MVs induced angiogenesis through p38 activation and angiogenesis induced by CD133-containing MVs was insensitive to the anti-vascular endothelial growth factor antibody bevacizumab. Survival analysis revealed that high expression level of CD133 correlated with poor prognosis in patients with metastatic CRC. These findings suggest that CD133-containing MVs act as key regulators of angiogenesis and are related to the prognosis of CRC patients.

17.
J Biol Chem ; 286(49): 42072-42081, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22009750

RESUMEN

Extracellular proteolysis mediates tissue homeostasis. In cancer, altered proteolysis leads to abnormal tumor growth, inflammation, tissue invasion, and metastasis. Matrix metalloproteinase-9 (MMP-9) represents one of the most prominent proteinases associated with inflammation and tumorigenesis. The recently identified human transcription factor sLZIP is a member of the leucine zipper transcription factor family. Although sLZIP is known to function in ligand-induced transactivation of the glucocorticoid receptor, its exact functions and target genes are not known. In this study, we investigated the role of sLZIP in MMP-9 expression and its involvement in cervical cancer development. Our results show that sLZIP increased the expression of MMP-9 at both the mRNA and protein levels and the proteolytic activity of MMP-9 in HeLa and SiHa cells. sLZIP also increased the transcriptional activity of MMP-9 by binding directly to the cAMP-responsive element of the MMP-9 promoter region. Involvement of sLZIP in MMP-9 expression was further supported by the fact that ME-180 cells expressing sLZIP siRNA were refractory to MMP-9 expression. Results from wound healing and invasion assays showed that sLZIP enhanced both the migration and invasion of cervical cancer cells. The increased migration and invasion of HeLa and SiHa cells that were induced by sLZIP were abrogated by inhibition of the proteolytic activity of MMP-9. These results indicate that sLZIP plays a critical role in MMP-9 expression and is probably involved in invasion and metastasis of cervical cancer.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/química , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 9 de la Matriz/biosíntesis , Neoplasias del Cuello Uterino/metabolismo , Línea Celular Tumoral , Movimiento Celular , Femenino , Células HeLa , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Regiones Promotoras Genéticas , Proteolisis , Interferencia de ARN
18.
J Cell Physiol ; 227(6): 2567-77, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21898402

RESUMEN

In this study, we investigated the effects of CCL2 on constitutive apoptosis of normal and asthmatic neutrophils. CCL2 blocked the constitutive apoptosis of normal neutrophils through CCR2. CCL2 also induced elevation of the cytosolic Ca(2+) concentration but had no effect on normal neutrophil chemotaxis. Constitutive apoptosis, calcium influx, and cell migration of asthmatic neutrophils were not affected by CCL2 stimulation. Supernatant collected from CCL2-treated normal neutrophils inhibited the constitutive apoptosis of normal neutrophils. Anti-apoptotic signaling mediated by CCL2 was found to be associated with the PI3K/Akt/ERK/NF-κB cascade in normal neutrophils. Both the cleavage of procaspase 3 and procaspase 9 and the decrease of in Mcl-1 expression were delayed by CCL2 stimulation. Inhibition of NF-κB blocked constitutive apoptosis of neutrophils from asthmatic patients via inhibition of the cleavage of procaspase 3 and procaspase 9, in contrast to normal neutrophils. NF-κB was involved in CCL2-induced anti-apoptotic signaling in normal neutrophils, whereas NF-κB functioned as a basal pro-apoptotic factor in asthmatic neutrophils. A better understanding of the difference in the regulation of neutrophil apoptosis due to CCL2 between normal individuals and asthmatics will enable elucidation of the role of CC chemokine in neutrophils and a framework for understanding the pathogenesis of asthma.


Asunto(s)
Apoptosis , Asma/inmunología , Quimiocina CCL2/metabolismo , Neutrófilos/inmunología , Adulto , Apoptosis/efectos de los fármacos , Asma/patología , Estudios de Casos y Controles , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Persona de Mediana Edad , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , FN-kappa B/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores CCR2/metabolismo , Transducción de Señal , Factores de Tiempo , Adulto Joven
19.
J Cell Biochem ; 113(9): 2845-55, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22488409

RESUMEN

Synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) has been reported to exhibit anti-invasive and anti-metastatic activities by suppressing the enzymatic activity of matrix metalloproteinase (MMP)-9, but the underlying mechanism remains unclear. Here, we show that 4-HPR blocks the activity of MMP-9 in two ways: by reducing phorbol 12-myristate 13-acetate (PMA)-induced MMP-9 secretion and by suppressing cell invasion through the downregulation of MMP-9 gene transcription in MCF-7 breast cancer cells. 4-HPR inhibits the transcriptional activity of MMP-9 by reducing the DNA-binding activity of NF-κB on the MMP-9 promoter as well as by inhibiting the degradation of IκBα, leading to cytoplasmic accumulation of NF-κB. We also found that 4-HPR inhibits invasion and MMP-9 expression in the highly metastatic breast cancer cell line MDA-MB-231. Thus, 4-HPR might be a potent anti-invasive agent that works by suppressing MMP-9 expression via the NF-κB signaling pathway.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fenretinida/farmacología , Metaloproteinasa 9 de la Matriz/metabolismo , FN-kappa B/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Immunoblotting , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Acetato de Tetradecanoilforbol/farmacología
20.
Biochem Biophys Res Commun ; 423(1): 188-94, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22640735

RESUMEN

Matrix metalloproteinase-9 (MMP-9) plays an important role in the invasion and metastasis of cancer cells. However, the regulatory mechanism of MMP-9 expression and its biological effects on breast cancer development remain obscure. In the current study, we examined the potential role of annexin A1 (ANXA1) in regulating migration and invasion in breast cancer cell lines. Both ANXA1 mRNA and protein are expressed in the highly invasive, hormone-insensitive human breast cancer cell lines MDA-MB-231 and SKBr3, but not in the hormone-responsive cell lines MCF-7 and T47D. Downregulation of ANXA1 expression with specific small interfering RNAs (ANXA1 siRNA) in MDA-MB-231 cells resulted in decreased cancer cell migration and invasion. Ablation of ANXA1 expression decreases the expression of MMP-9 at both the mRNA and protein levels and also reduces the proteolytic activity of MMP-9 in MDA-MB-231 cells. Moreover, silencing ANXA1 also decreases the transcriptional activity of MMP-9 by the suppression of nuclear factor kappa-B (NF-κB) activity. Collectively, these results indicate that ANXA1 functions as a positive regulator of MMP-9 expression and invasion of breast cancer cells through specific activation of the NF-κB signaling pathway.


Asunto(s)
Anexina A1/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 9 de la Matriz/genética , Anexina A1/genética , Línea Celular Tumoral , ADN/metabolismo , Femenino , Humanos , FN-kappa B/metabolismo , Invasividad Neoplásica , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA