Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Sci ; 110(9): 2783-2793, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31325403

RESUMEN

Oral cancer, a subtype of head and neck cancer, is characterized by increased infiltrating regulatory T cells (Treg); however, the pathological significance of the increase in Tregs in disease prognosis and progression and their underlying mechanism remain unestablished. C-C motif chemokine ligand 22 (CCL22) has been implicated in the recruitment of Tregs. We used RT-qPCR to determine CCL22 mRNA expression in clinical specimens and cultured cells. Loss-of-function and gain-of-function studies were carried out to analyze the effects of CCL22 modulations on cell proliferation, migration, invasion, and tumorigenesis and the mechanism involved in the deregulation of CCL22. In oral cancer specimens, CCL22 mRNA was upregulated. The increase was not only associated with reduced disease-free survival but also strongly correlated with an increase in FOXP3 mRNA, a master regulator of Treg development and functions. Silencing CCL22 expression reduced cell proliferation, migration, and invasion, whereas ectopic overexpression showed opposite effects. Manipulation of CCL22 expression in cancer cells altered tumorigenesis in both immune-compromised and -competent mice, supporting both autonomous and non-autonomous actions of CCL22. Release of interleukin 1ß (IL-1ß) from cancer-associated fibroblasts (CAF) induces CCL22 mRNA expression in oral cancer cells by activating transcription factor nuclear factor kappa B (NF-κB). Our data support a model in which CAF-derived IL-1ß, CCL22, and its receptor CCR4 foster a protumor environment by promoting cell transformation and Treg infiltration. Intervention of the IL-1ß-CCL22-CCR4 signaling axis may offer a novel therapeutic strategy for oral cancer treatment.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Quimiocina CCL22/metabolismo , Interleucina-1beta/metabolismo , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Animales , Fibroblastos Asociados al Cáncer/inmunología , Línea Celular Tumoral , Movimiento Celular/inmunología , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/patología , Quimiocina CCL22/genética , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-1beta/genética , Masculino , Ratones , Persona de Mediana Edad , Mucosa Bucal/patología , Mucosa Bucal/cirugía , Neoplasias de la Boca/inmunología , Neoplasias de la Boca/mortalidad , Neoplasias de la Boca/cirugía , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/patología , Pronóstico , ARN Interferente Pequeño/metabolismo , Receptores CCR4/metabolismo , Transducción de Señal/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Carcinoma de Células Escamosas de Cabeza y Cuello/cirugía , Análisis de Supervivencia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Biol Chem ; 292(5): 1637-1647, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-27998979

RESUMEN

Glucocorticoids are a general class of steroids that possess renoprotective activity in glomeruli through their interaction with the glucocorticoid receptor. However, the mechanisms by which glucocorticoids ameliorate proteinuria and glomerular disease are not well understood. In this study, we demonstrated that α actinin 4 (ACTN4), an actin-cross-linking protein known to coordinate cytoskeletal organization, interacts with the glucocorticoid receptor (GR) in the nucleus of human podocytes (HPCs), a key cell type in the glomerulus critical for kidney filtration function. The GR-ACTN4 complex enhances glucocorticoid response element (GRE)-driven reporter activity. Stable knockdown of ACTN4 by shRNA in HPCs significantly reduces dexamethasone-mediated induction of GR target genes and GRE-driven reporter activity without disrupting dexamethasone-induced nuclear translocation of GR. Synonymous mutations or protein expression losses in ACTN4 are associated with kidney diseases, including focal segmental glomerulosclerosis, characterized by proteinuria and podocyte injury. We found that focal segmental glomerulosclerosis-linked ACTN4 mutants lose their ability to bind liganded GR and support GRE-mediated transcriptional activity. Mechanistically, GR and ACTN4 interact in the nucleus of HPCs. Furthermore, disruption of the LXXLL nuclear receptor-interacting motif present in ACTN4 results in reduced GR interaction and dexamethasone-mediated transactivation of a GRE reporter while still maintaining its actin-binding activity. In contrast, an ACTN4 isoform, ACTN4 (Iso), that loses its actin-binding domain is still capable of potentiating a GRE reporter. Dexamethasone induces the recruitment of ACTN4 and GR to putative GREs in dexamethasone-transactivated promoters, SERPINE1, ANGPLT4, CCL20, and SAA1 as well as the NF-κB (p65) binding sites on GR-transrepressed promoters such as IL-1ß, IL-6, and IL-8 Taken together, our data establish ACTN4 as a transcriptional co-regulator that modulates both dexamethasone-transactivated and -transrepressed genes in podocytes.


Asunto(s)
Actinina/biosíntesis , Dexametasona/farmacología , Podocitos/metabolismo , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta/fisiología , Activación Transcripcional/efectos de los fármacos , Actinina/genética , Citocinas/biosíntesis , Citocinas/genética , Células HEK293 , Células HeLa , Humanos , Podocitos/citología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/genética
3.
J Biol Chem ; 292(24): 10048-10060, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28432122

RESUMEN

IFNs are effective in inhibiting angiogenesis in preclinical models and in treating several angioproliferative disorders. However, the detailed mechanisms of IFNα-mediated anti-angiogenesis are not completely understood. Stat1/2/3 and PML are IFNα downstream effectors and are pivotal regulators of angiogenesis. Here, we investigated PML's role in the regulation of Stat1/2/3 activity. In Pml knock-out (KO) mice, ablation of Pml largely reduces IFNα angiostatic ability in Matrigel plug assays. This suggested an essential role for PML in IFNα's anti-angiogenic function. We also demonstrated that PML shared a large cohort of regulatory genes with Stat1 and Stat3, indicating an important role of PML in regulating Stat1 and Stat3 activity. Using molecular tools and primary endothelial cells, we demonstrated that PML positively regulates Stat1 and Stat2 isgylation, a ubiquitination-like protein modification. Accordingly, manipulation of the isgylation system by knocking down USP18 altered IFNα-PML axis-mediated inhibition of endothelial cell migration and network formation. Furthermore, PML promotes turnover of nuclear Stat3, and knockdown of PML mitigates the effect of LLL12, a selective Stat3 inhibitor, on IFNα-mediated anti-angiogenic activity. Taken together, we elucidated an unappreciated mechanism in which PML, an IFNα-inducible effector, possess potent angiostatic activity, doing so in part by forming a positive feedforward loop with Stat1/2 and a negative feedback loop with Stat3. The interplay between PML, Stat1/Stat2, and Stat3 contributes to IFNα-mediated inhibition of angiogenesis, and disruption of this network results in aberrant IFNα signaling and altered angiostatic activity.


Asunto(s)
Endotelio Vascular/metabolismo , Interferón-alfa/metabolismo , Neovascularización Patológica/prevención & control , Proteína de la Leucemia Promielocítica/metabolismo , Factor de Transcripción STAT1/agonistas , Factor de Transcripción STAT2/agonistas , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Línea Celular , Células Cultivadas , Endopeptidasas/química , Endopeptidasas/genética , Endopeptidasas/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interferón-alfa/genética , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Fisiológica , Proteína de la Leucemia Promielocítica/antagonistas & inhibidores , Proteína de la Leucemia Promielocítica/genética , Procesamiento Proteico-Postraduccional , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
4.
J Biol Chem ; 291(11): 5512-5526, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26786102

RESUMEN

The transcription factor c-Fos controls many important cellular processes, including cell growth and apoptosis. c-Fos expression is rapidly elevated in the prostate upon castration-mediated androgen withdrawal through an undefined mechanism. Here we show that androgens (5α-dihydrotestosterone and R1881) suppress c-Fos protein and mRNA expression induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) or EGF in human prostate cancer (PCa) cell lines. Such suppression transpires through a transcriptional mechanism, predominantly at the proximal serum response element of the c-fos promoter. We show that androgen signaling suppresses TPA-induced c-Fos expression through repressing a PKC/MEK/ERK/ELK-1 signaling pathway. Moreover, our results support the hypothesis that p38(MAPK), PI3K, and PKCδ are involved in the androgenic regulation of c-Fos through controlling MEK/ERK. Stable silencing of c-Fos and PKCδ with shRNAs suggests that R1881 promotes cell death induced by low-dose TPA through a mechanism that is dependent on both PKCδ and loss of c-Fos expression. Reciprocally, loss of either PKCδ or c-Fos activates p38(MAPK) while suppressing the activation of ERK1/2. We also provide the first demonstration that R1881 permits cell death induced by low-dose TPA in the LNCaP androgen-dependent PCa cell line and that TPA-induced cell death is independent of exogenous androgen in the castration-resistant variants of LNCaP, C4-2 and C4-2B. Acquisition of androgen-independent killing by TPA correlates with activation of p38(MAPK), suppression of ERK1/2, and loss of c-Fos. These results provide new insights into androgenic control of c-Fos and use of PKC inhibitors in PCa therapy.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Andrógenos/farmacología , Dihidrotestosterona/farmacología , Metribolona/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-fos/genética , Acetato de Tetradecanoilforbol/farmacología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Próstata/efectos de los fármacos , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteína Quinasa C/metabolismo , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
J Biol Chem ; 290(1): 338-49, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25411248

RESUMEN

Glomerular podocytes are highly specialized terminally differentiated cells that act as a filtration barrier in the kidney. Mutations in the actin-binding protein, α-actinin 4 (ACTN4), are linked to focal segmental glomerulosclerosis (FSGS), a chronic kidney disease characterized by proteinuria. Aberrant activation of NF-κB pathway in podocytes is implicated in glomerular diseases including proteinuria. We demonstrate here that stable knockdown of ACTN4 in podocytes significantly reduces TNFα-mediated induction of NF-κB target genes, including IL-1ß and NPHS1, and activation of an NF-κB-driven reporter without interfering with p65 nuclear translocation. Overexpression of ACTN4 and an actin binding-defective variant increases the reporter activity. In contrast, an FSGS-linked ACTN4 mutant, K255E, which has increased actin binding activity and is predominantly cytoplasmic, fails to potentiate NF-κB activity. Mechanistically, IκBα blocks the association of ACTN4 and p65 in the cytosol. In response to TNFα, both NF-κB subunits p65 and p50 translocate to the nucleus, where they bind and recruit ACTN4 to their targeted promoters, IL-1ß and IL-8. Taken together, our data identify ACTN4 as a novel coactivator for NF-κB transcription factors in podocytes. Importantly, this nuclear function of ACTN4 is independent of its actin binding activity in the cytoplasm.


Asunto(s)
Actinina/genética , FN-kappa B/genética , Podocitos/metabolismo , Transcripción Genética , Actinina/antagonistas & inhibidores , Actinina/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Línea Celular Transformada , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Podocitos/citología , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
6.
J Biol Chem ; 288(35): 25375-25386, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23861398

RESUMEN

Cytokine modulation of the endothelium is considered an important contributor to the inflammation response. TNFα is an early response gene during the initiation of inflammation. However, the detailed mechanism by which TNFα induces proinflammatory gene expression is not completely understood. In this report, we demonstrate that silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) represses the expression of a subset of TNFα target genes in human umbilical vein endothelial cells. Upon TNFα stimulation, we observed an increase in the E3 ubiquitin ligase ß-TrCP1 and a decrease in SMRT protein levels. We show that ß-TrCP1 interacts with SMRT in a phosphorylation-independent manner and cooperates with the E2 ubiquitin-conjugating enzyme E2D2 to promote ubiquitination-dependent SMRT degradation. Knockdown of ß-TrCP1 increases SMRT protein accumulation, increases SMRT association with its targeted promoters, and decreases SMRT target gene expression. Taken together, our results support a model in which TNFα-induced ß-TrCP1 accumulation promotes SMRT degradation and the subsequent induction of proinflammatory gene expression.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Modelos Biológicos , Co-Represor 2 de Receptor Nuclear/metabolismo , Proteolisis , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas con Repetición de beta-Transducina/metabolismo , Células HEK293 , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Co-Represor 2 de Receptor Nuclear/genética , Fosforilación/fisiología , Factor de Necrosis Tumoral alfa/genética , Ubiquitina/genética , Ubiquitina/metabolismo , Proteínas con Repetición de beta-Transducina/genética
7.
J Biol Chem ; 288(41): 29746-59, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-23986437

RESUMEN

The promyelocytic leukemia protein is a well known tumor suppressor, but its role in metabolism is largely unknown. Mice with a deletion in the gene for PML (KO mice) exhibit altered gene expression in liver, adipose tissue, and skeletal muscle, an accelerated rate of fatty acid metabolism, abnormal glucose metabolism, constitutive AMP-activating kinase (AMPK) activation, and insulin resistance in skeletal muscle. Last, an increased rate of energy expenditure protects PML KO mice from the effects of obesity induced by a Western diet. Collectively, our study uncovers a previously unappreciated role of PML in the regulation of metabolism and energy balance in mice.


Asunto(s)
Metabolismo Energético/genética , Proteínas Nucleares/genética , Obesidad/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipoquinas/genética , Tejido Adiposo/metabolismo , Animales , Western Blotting , Temperatura Corporal/genética , Antígenos CD36/genética , Dieta/efectos adversos , Ácidos Grasos/metabolismo , Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Hígado/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Músculo Esquelético/metabolismo , Proteínas Nucleares/deficiencia , Obesidad/etiología , Obesidad/metabolismo , Oxidación-Reducción , Proteína de la Leucemia Promielocítica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/deficiencia , Proteínas Supresoras de Tumor/deficiencia
8.
Cell Death Differ ; 31(6): 768-778, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38627584

RESUMEN

The alternative splicing of PML precursor mRNA gives rise to various PML isoforms, yet their expression profile in breast cancer cells remains uncharted. We discovered that PML1 is the most abundant isoform in all breast cancer subtypes, and its expression is associated with unfavorable prognosis in estrogen receptor-positive (ER+) breast cancers. PML depletion reduces cell proliferation, invasion, and stemness, while heterologous PML1 expression augments these processes and fuels tumor growth and resistance to fulvestrant, an FDA-approved drug for ER+ breast cancer, in a mouse model. Moreover, PML1, rather than the well-known tumor suppressor isoform PML4, rescues the proliferation of PML knockdown cells. ChIP-seq analysis reveals significant overlap between PML-, ER-, and Myc-bound promoters, suggesting their coordinated regulation of target gene expression, including genes involved in breast cancer stem cells (BCSCs), such as JAG1, KLF4, YAP1, SNAI1, and MYC. Loss of PML reduces BCSC-related gene expression, and exogenous PML1 expression elevates their expression. Consistently, PML1 restores the association of PML with these promoters in PML-depleted cells. We identified a novel association between PML1 and WDR5, a key component of H3K4 methyltransferase (HMTs) complexes that catalyze H3K4me1 and H3K4me3. ChIP-seq analyses showed that the loss of PML1 reduces H3K4me3 in numerous loci, including BCSC-associated gene promoters. Additionally, PML1, not PML4, re-establishes the H3K4me3 mark on these promoters in PML-depleted cells. Significantly, PML1 is essential for recruiting WDR5, MLL1, and MLL2 to these gene promoters. Inactivating WDR5 by knockdown or inhibitors phenocopies the effects of PML1 loss, reducing BCSC-related gene expression and tumorsphere formation and enhancing fulvestrant's anticancer activity. Our findings challenge the conventional understanding of PML as a tumor suppressor, redefine its role as a promoter of tumor growth in breast cancer, and offer new insights into the unique roles of PML isoforms in breast cancer.


Asunto(s)
Neoplasias de la Mama , Histonas , Factor 4 Similar a Kruppel , Células Madre Neoplásicas , Proteína de la Leucemia Promielocítica , Receptores de Estrógenos , Humanos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Femenino , Proteína de la Leucemia Promielocítica/metabolismo , Proteína de la Leucemia Promielocítica/genética , Animales , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica
9.
J Biol Chem ; 287(28): 23356-67, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22589541

RESUMEN

Promyelocytic leukemia protein (PML) is a tumor suppressor that is highly expressed in vascular endothelium and inflamed tissues, yet its role in inflammation-associated cytokine-regulated angiogenesis and underlying mechanism remains largely unclear. We show that tumor necrosis factor α (TNFα) and interferon α (IFNα) stimulate PML expression while suppressing EC network formation and migration, two key events during angiogenesis. By a knockdown approach, we demonstrate that PML is indispensable for TNFα- and IFNα-mediated inhibition of EC network formation. We further demonstrate that signal transducer and activator of transcription 1 (STAT1) binds PML promoter and that is an important regulator of PML expression. Knockdown of STAT1 reduces endogenous PML and blocks TNFα- and IFNα-induced PML accumulation and relieves TNFα- and IFNα-mediated inhibition of EC network formation. Our data also indicate that PML regulates EC migration, in part, by modulating expression of downstream genes, such as negatively regulating integrin ß1 (ITGB1). In addition, knockdown of STAT1 or PML alleviates TNFα- and IFNα-mediated inhibition of ITGB1 expression. Antibody blockade demonstrates that ITGB1 is functionally important for PML- and STAT1-regulated EC migration. Taken together, our data provide novel mechanistic insights that PML functions as a negative regulator in EC network formation and migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Interferón-alfa/farmacología , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Supresoras de Tumor/metabolismo , Células Cultivadas , Células Endoteliales/citología , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Microscopía Fluorescente , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína de la Leucemia Promielocítica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
10.
J Biol Chem ; 287(42): 35418-35429, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22908231

RESUMEN

α-Actinins (ACTNs) are a family of proteins cross-linking actin filaments that maintain cytoskeletal organization and cell motility. Recently, it has also become clear that ACTN4 can function in the nucleus. In this report, we found that ACTN4 (full length) and its spliced isoform ACTN4 (Iso) possess an unusual LXXLL nuclear receptor interacting motif. Both ACTN4 (full length) and ACTN4 (Iso) potentiate basal transcription activity and directly interact with estrogen receptor α, although ACTN4 (Iso) binds ERα more strongly. We have also found that both ACTN4 (full length) and ACTN4 (Iso) interact with the ligand-independent and the ligand-dependent activation domains of estrogen receptor α. Although ACTN4 (Iso) interacts efficiently with transcriptional co-activators such as p300/CBP-associated factor (PCAF) and steroid receptor co-activator 1 (SRC-1), the full length ACTN4 protein either does not or does so weakly. More importantly, the flanking sequences of the LXXLL motif are important not only for interacting with nuclear receptors but also for the association with co-activators. Taken together, we have identified a novel extended LXXLL motif that is critical for interactions with both receptors and co-activators. This motif functions more efficiently in a spliced isoform of ACTN4 than it does in the full-length protein.


Asunto(s)
Actinina/metabolismo , Empalme Alternativo/fisiología , Receptor alfa de Estrógeno/metabolismo , Coactivador 1 de Receptor Nuclear/metabolismo , Transcripción Genética/fisiología , Factores de Transcripción p300-CBP/metabolismo , Actinina/genética , Secuencias de Aminoácidos , Línea Celular Tumoral , Receptor alfa de Estrógeno/genética , Humanos , Coactivador 1 de Receptor Nuclear/genética , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción p300-CBP/genética
11.
J Biol Chem ; 287(31): 25869-80, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22648458

RESUMEN

Histone acetylation was significantly increased in retinas from diabetic rats, and this acetylation was inhibited in diabetics treated with minocycline, a drug known to inhibit early diabetic retinopathy in animals. Histone acetylation and expression of inflammatory proteins that have been implicated in the pathogenesis of diabetic retinopathy were increased likewise in cultured retinal Müller glia grown in a diabetes-like concentration of glucose. Both the acetylation and induction of the inflammatory proteins in elevated glucose levels were significantly inhibited by inhibitors of histone acetyltransferase (garcinol and antisense against the histone acetylase, p300) or activators of histone deacetylase (theophylline and resveratrol) and were increased by the histone deacetylase inhibitor, suberolylanilide hydroxamic acid. We conclude that hyperglycemia causes acetylation of retinal histones (and probably other proteins) and that the acetylation contributes to the hyperglycemia-induced up-regulation of proinflammatory proteins and thereby to the development of diabetic retinopathy.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Minociclina/farmacología , Procesamiento Proteico-Postraduccional , Acetilación/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Línea Celular , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/enzimología , Retinopatía Diabética/metabolismo , Histona Acetiltransferasas/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Histonas/química , Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Minociclina/uso terapéutico , Datos de Secuencia Molecular , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fragmentos de Péptidos/química , Ratas , Ratas Endogámicas Lew , Retina/efectos de los fármacos , Retina/enzimología , Retina/metabolismo , Espectrometría de Masas en Tándem , Terpenos/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
J Biol Chem ; 287(15): 12027-35, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22351778

RESUMEN

Mutations in α-actinin 4 (ACTN4) are linked to familial forms of focal segmental glomerulosclerosis (FSGS), a kidney disease characterized by proteinuria due to podocyte injury. The mechanisms underlying ACTN4 mutant-associated FSGS are not completely understood. Although α-actinins are better known to cross-link actin filaments and modulate cytoskeletal organization, we have previously shown that ACTN4 interacts with transcription factors including estrogen receptor and MEF2s and potentiates their transcriptional activity. Nuclear receptors including retinoic acid receptor (RAR) have been proposed to play a protective role in podocytes. We show here that ACTN4 interacts with and enhances transcriptional activation by RARα. In addition, FSGS-linked ACTN4 mutants not only mislocalized to the cytoplasm, but also lost their ability to associate with nuclear receptors. Consequently, FSGS-linked ACTN4 mutants failed to potentiate transcriptional activation by nuclear hormone receptors in podocytes. In addition, overexpression of these mutants suppressed the transcriptional activity mediated by endogenous wild-type ACTN4 possibly by a cytoplasmic sequestration mechanism. Our data provide the first link between FSGS-linked ACTN4 mutants and transcriptional activation by nuclear receptor such as RARα and peroxisome proliferator-activated receptor γ.


Asunto(s)
Actinina/genética , Glomeruloesclerosis Focal y Segmentaria/genética , Mutación Missense , Receptores de Ácido Retinoico/metabolismo , Transcripción Genética , Actinina/metabolismo , Línea Celular , Humanos , Proteínas Mutantes/metabolismo , PPAR gamma/metabolismo , Podocitos/metabolismo , Unión Proteica , Transporte de Proteínas , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/genética , Proteínas Recombinantes/metabolismo , Receptor alfa de Ácido Retinoico , Activación Transcripcional , Tretinoina/fisiología
13.
Res Sq ; 2023 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-37720048

RESUMEN

The alternative splicing of PML precursor mRNA gives rise to various PML isoforms, yet their expression profile in breast cancer cells remains uncharted. We discovered that PML1 is the most abundant isoform in all breast cancer subtypes, and its expression is associated with unfavorable prognosis in estrogen receptor-positive (ER+) breast cancers. PML depletion reduces cell proliferation, invasion, and stemness, while heterologous PML1 expression augments these processes and fuels tumor growth and resistance to fulvestrant, an FDA-approved drug for ER + breast cancer, in a mouse model. Moreover, PML1, rather than the well-known tumor suppressor isoform PML4, rescues the proliferation of PML knockdown cells. ChIP-seq analysis reveals significant overlap between PML-, ER-, and Myc-bound promoters, suggesting their coordinated regulation of target gene expression, including genes involved in breast cancer stem cells (BCSCs), such as JAG1, KLF4, YAP1, SNAI1, and MYC. Loss of PML reduces BCSC-related gene expression, and exogenous PML1 expression elevates their expression. Consistently, PML1 restores the association of PML with these promoters in PML-depleted cells. We identified a novel association between PML1 and WDR5, a key component of H3K4 methyltransferase (HMTs) complexes that catalyze H3K4me1 and H3K4me3. ChIP-seq analyses showed that the loss of PML1 reduces H3K4me3 in numerous loci, including BCSC-associated gene promoters. Additionally, PML1, not PML4, re-establishes the H3K4me3 mark on these promoters in PML-depleted cells. Significantly, PML1 is essential for recruiting WDR5, MLL1, and MLL2 to these gene promoters. Inactivating WDR5 by knockdown or inhibitors phenocopies the effects of PML1 loss, reducing BCSC-related gene expression and tumorsphere formation and enhancing fulvestrant's anticancer activity. Our findings challenge the conventional understanding of PML as a tumor suppressor, redefine its role as a promoter of tumor growth in breast cancer and offer new insights into the unique roles of PML isoforms in breast cancer.

14.
J Biol Chem ; 286(52): 44403-11, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22033920

RESUMEN

The promyelocytic leukemia (PML) protein is a tumor suppressor that has an important role in several cellular processes, including apoptosis, viral infection, DNA damage repair, cell cycle regulation, and senescence. PML is an essential component of sub-nuclear structures called PML nuclear bodies (NBs). Our laboratory has previously demonstrated that the peptidyl-prolyl cis-trans isomerase, Pin1, binds and targets PML for degradation in a phosphorylation-dependent manner. To further elucidate the mechanisms underlying Pin1-mediated PML degradation, we aimed to identify one or more factors that promote PML phosphorylation. Here we show that treatment with U0126, an inhibitor of the ERK2 upstream kinases MEK1/2, leads to an increase in PML protein accumulation and an inhibition of the interaction between Pin1 and PML in MDA-MB-231 breast cancer cells. Consistent with this observation, phosphorylated ERK2 partially co-localized with PML NBs. Although U0126 up-regulated exogenous wild-type PML levels, it did not have an effect on the steady-state level of a mutant form of PML that is defective in binding Pin1. In addition, exogenous wild-type, but not Pin1 binding-defective PML protein expression levels were decreased by overexpression of ERK2. In contrast, knockdown of ERK2 by siRNA resulted in an increase in PML protein levels and an increase in the formation of PML NBs. Using phospho-specific antibodies, we identified Ser-403 and Ser-505 as the ERK2 targets that promote Pin1-mediated PML degradation. Finally, we demonstrated that EGF induced activation of ERK and interaction between PML and phosphorylated ERK resulting in a decrease in PML protein levels. Taken together, our results support a model in which Pin1 promotes PML degradation in an ERK2-dependent manner.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Modelos Biológicos , Proteínas Nucleares/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Proteolisis , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Butadienos/farmacología , Inhibidores Enzimáticos/farmacología , Células HEK293 , Células HeLa , Humanos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Peptidilprolil Isomerasa de Interacción con NIMA , Nitrilos/farmacología , Proteínas Nucleares/genética , Isomerasa de Peptidilprolil/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteína de la Leucemia Promielocítica , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
15.
J Biol Chem ; 286(3): 1850-9, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21078666

RESUMEN

Alpha actinins (ACTNs) are known for their ability to modulate cytoskeletal organization and cell motility by cross-linking actin filaments. We show here that ACTN4 harbors a functional LXXLL receptor interaction motif, interacts with nuclear receptors in vitro and in mammalian cells, and potently activates transcription mediated by nuclear receptors. Whereas overexpression of ACTN4 potentiates estrogen receptor α (ERα)-mediated transcription in transient transfection reporter assays, knockdown of ACTN4 decreases it. In contrast, histone deacetylase 7 (HDAC7) inhibits estrogen receptor α (ERα)-mediated transcription. Moreover, the ACTN4 mutant lacking the CaM (calmodulin)-like domain that is required for its interaction with HDAC7 fails to activate transcription by ERα. Chromatin immunoprecipitation (ChIP) assays demonstrate that maximal associations of ACTN4 and HDAC7 with the pS2 promoter are mutually exclusive. Knockdown of ACTN4 significantly decreases the expression of ERα target genes including pS2 and PR and also affects cell proliferation of MCF-7 breast cancer cells with or without hormone, whereas knockdown of HDAC7 exhibits opposite effects. Interestingly, overexpression of wild-type ACTN4, but not the mutants defective in interacting with ERα or HDAC7, results in an increase in pS2 and PR mRNA accumulation in a hormone-dependent manner. In summary, we have identified ACTN4 as a novel, atypical coactivator that regulates transcription networks to control cell growth.


Asunto(s)
Actinina/metabolismo , Neoplasias de la Mama/metabolismo , Proliferación Celular , Receptor alfa de Estrógeno/metabolismo , Histona Desacetilasas/metabolismo , Proteínas de Neoplasias/metabolismo , Actinina/genética , Secuencias de Aminoácidos , Neoplasias de la Mama/genética , Línea Celular Tumoral , Receptor alfa de Estrógeno/genética , Femenino , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Humanos , Proteínas de Neoplasias/genética , Presenilina-2/biosíntesis , Presenilina-2/genética , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína
16.
BMC Genomics ; 13: 453, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22947142

RESUMEN

BACKGROUND: Promyelocytic leukemia protein (PML) is a tumor suppressor that is highly expressed in endothelial cells nonetheless its role in endothelial cell biology remains elusive. Tumor necrosis factor alpha (TNFα) is an important cytokine associated with many inflammation-related diseases. We have previously demonstrated that TNFα induces PML protein accumulation. We hypothesized that PML may play a role in TNFα signaling pathway. To identify potential PML target genes and investigate the putative crosstalk between PML's function and TNFα signaling in endothelial cells, we carried out a microarray analysis in human primary umbilical endothelial cells (HUVECs). RESULTS: We found that PML and TNFα regulate common and distinct genes involved in a similar spectrum of biological processes, pathways and human diseases. More importantly, we found that PML is required for fine-tuning of TNFα-mediated immune and inflammatory responses. Furthermore, our data suggest that PML and TNFα synergistically regulate cell adhesion by engaging multiple molecular mechanisms. Our biological functional assays exemplified that adhesion of U937 human leukocytes to HUVECs is co-regulated by PML and TNFα signaling. CONCLUSIONS: Together, our study identified PML as an essential regulator of TNFα signaling by revealing the crosstalk between PML knockdown-mediated effects and TNFα-elicited signaling, thereby providing novel insights into TNFα signaling in endothelial cells.


Asunto(s)
Regulación de la Expresión Génica/genética , Proteínas Nucleares/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adhesión Celular/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Análisis por Micromatrices , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/genética , Proteínas Supresoras de Tumor/genética , Células U937
17.
J Biol Chem ; 285(13): 9485-9492, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20100838

RESUMEN

Promyelocytic leukemia protein (PML) was originally identified as part of a chromosomal translocation that contributes to the development of acute promyelocytic leukemia (APL). Since its discovery, PML has been found to play diverse roles in different cellular processes. Notably, PML has anti-proliferative and pro-apoptotic activity that supports its role as a tumor suppressor. We have previously shown that the peptidyl-prolyl isomerase Pin1 is able to affect cell proliferation and hydrogen peroxide (H(2)O(2))-mediated cell death through modulation of the steady-state levels of PML. We have extended these studies to show that the interaction between PML and Pin1 is targeted by multiple extracellular signals in the cell. We show that H(2)O(2) up-regulates and IGF-1 down-regulates PML expression in a Pin1-dependent manner. Interestingly, we found that H(2)O(2)- and IGF-1-mediated alteration in PML accumulation regulate MDA-MB-231 cell migration. Furthermore, we show that the control of cell migration by PML, and thus H(2)O(2) and IGF-1, results from PML-dependent decreased expression of integrin beta1 (ITGB1). Knockdown of Pin1 leads to decreased cell migration, lower levels of ITGB1 expression and resistance to IGF-1- and H(2)O(2)-induced changes in cell migration and ITGB1 expression. Taken together, our work identifies PML as a common target for H(2)O(2) and IGF-1 and supports a novel tumor suppressive role for PML in controlling cell migration through the expression of ITGB1.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Peróxido de Hidrógeno/química , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Línea Celular Tumoral , Movimiento Celular , Regulación hacia Abajo , Humanos , Integrina beta1/metabolismo , Microscopía Fluorescente/métodos , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/química , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Cicatrización de Heridas
18.
Biochim Biophys Acta ; 1803(10): 1186-97, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20621129

RESUMEN

Class IIa HDACs including HDAC7 play a role in gene expression, cell differentiation, and animal development through their association with transcription factors such as myogenic enhancer factors 2 (MEF2s). In this study, we show that endogenous HDAC7 localizes to both the nucleus and the cytoplasm of C2C12 myoblasts but is exclusively retained in the cytoplasm of myotubes after completion of differentiation process. To elucidate the role of differential distribution of HDAC7 during myogenesis, we examined the effects of stably expressed HDAC7 mutants on myogenesis. Expression of nuclear-retained HDAC7 mutants significantly inhibits myogenesis in C2C12 cells and reduces the expression of muscle-specific myosin heavy chain (MHC) and myogenin. The inhibition in myocyte differentiation can be partially relieved by introduction of a mutation disrupting HDAC7:MEF2 interaction. Since phosphorylation of HDAC7 plays an important role in its nucleocytoplasmic shuttling, we further investigated the expression and distribution of phosphorylated HDAC7. To our surprise, the phosphorylation levels of HDAC7 at S344 and S479 were slightly decreased upon differentiation, whereas the phosphorylation of S178 was unchanged. Interestingly, a significant fraction of pS344- and/or pS479-HDAC7 localized to plasma membrane of myotubes. In addition, Ser178-phosphorylated (pS178) HDAC7 displays a predominantly actin filament-like structure before muscle differentiation. Consistent with this notion, HDAC7 partially colocalized with actin filaments; in particular, pS178-HDAC7 largely colocalized with actin filaments as indicated by phalloidin counter staining in myocytes. Furthermore, C2C12 cells expressing nuclear-retained HDAC7 display defects in migration. Our results provide novel insight into the mechanisms that regulate myocyte differentiation and migration by controlling the subcellular distribution of HDAC7 in differentiating myoblasts.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Histona Desacetilasas/metabolismo , Mioblastos/metabolismo , Actinas/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Movimiento Celular/genética , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Histona Desacetilasas/genética , Immunoblotting , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Microscopía Confocal , Mutación , Mioblastos/citología , Miogenina/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Fosforilación , Serina/genética , Serina/metabolismo
19.
J Biol Chem ; 284(52): 36395-36404, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19858209

RESUMEN

We have identified G protein suppressor 2 (GPS2) as a stable component of the SMRT corepressor complexes. GPS2 potently represses basal transcription, with the repression domain mapped to the N-terminal silencing mediator of retinoic acid and thyroid hormone receptor (SMRT)-interacting domain. Knockdown of GPS2 abrogates, whereas overexpression potentiates, SMRT-mediated repression activity. The SMRT complexes are involved in 4-hydroxyl-tamoxifen (4OHT)-mediated gene repression by estrogen receptor alpha (ERalpha). We show that 4OHT recruits SMRT and GPS2 to the promoter of pS2, an ERalpha target gene, in a dynamic manner. Unexpectedly, we also found that estradiol (E2) promotes promoter recruitment of the SMRT complexes. While knockdown of GPS2 compromised 4OHT-mediated repression, it enhanced E2-induced expression of a reporter gene and several endogenous ERalpha target genes, including pS2, cyclin D1 (CCND1), progesterone receptor (PR), and c-MYC. Finally, we show that depletion of GPS2 or SMRT by siRNA promotes cell proliferation in MCF-7 breast cancer cells. Thus, we concluded that GPS2 is an integral component of the SMRT complexes, important for ligand-dependent gene regulations by ERalpha and a suppressor for MCF-7 cell proliferation.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética/fisiología , Ciclina D1/biosíntesis , Ciclina D1/genética , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/genética , Estrógenos/farmacología , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , Estructura Terciaria de Proteína/fisiología , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Receptores de Progesterona/biosíntesis , Receptores de Progesterona/genética , Proteínas Represoras/genética , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Transcripción Genética/efectos de los fármacos
20.
Cancers (Basel) ; 12(6)2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-32471132

RESUMEN

The TGF-ß type III receptor (TGFBR3) is an essential constituent of the TGF-ß signaling. In this study, we observed a down-regulation of TGFBR3 in oral cancer, a subtype of head and neck cancer (HNC), and patients with low TGFBR3 had poor clinical outcomes. Ectopic expression of TGFBR3 decreased migration and invasion of oral cancer cells and lymph node metastasis of tumors, whereas depletion of TGFBR3 had the opposite effect. In SMAD4-positive OC-2 oral cancer cells, TGFBR3-mediated suppression requires both of its cytoplasmic interacting partners ARRB2 and GIPC1. We demonstrated that TGFBR3 induces the abundance of secreted angiogenin (ANG), a known pro-angiogenic factor, and ANG is essential and sufficient to mediate TGFBR3-dependent inhibition of migration and invasion of oral cancer cells. Notably, in SMAD4-deficient CAL-27 oral cancer cells, only GIPC1 is essential for TGFBR3-induced suppressive activity. Accordingly, HNC patients with low expressions of both TGFBR3 and GIPC1 had the poorest overall survival. In summary, we conclude that TGFBR3 is as a tumor suppressor via SMAD4-dependent and -independent manner in both tumor and stromal cells during oral carcinogenesis. Our study should facilitate the possibility of using TGFBR3-mediated tumor suppression for HNC treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA