Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Eur J Cell Biol ; 102(3): 151341, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37459799

RESUMEN

ING1 is a chromatin targeting subunit of the Sin3a histone deacetylase (HDAC) complex that alters chromatin structure to subsequently regulate gene expression. We find that ING1 knockdown increases expression of Twist1, Zeb 1&2, Snai1, Bmi1 and TSHZ1 drivers of EMT, promoting EMT and cell motility. ING1 expression had the opposite effect, promoting epithelial cell morphology and inhibiting basal and TGF-ß-induced motility in 3D organoid cultures. ING1 binds the Twist1 promoter and Twist1 was largely responsible for the ability of ING1 to reduce cell migration. Consistent with ING1 inhibiting Twist1 expression in vivo, an inverse relationship between ING1 and Twist1 levels was seen in breast cancer samples from The Cancer Genome Atlas (TCGA). The HDAC inhibitor vorinostat is approved for treatment of multiple myeloma and cutaneous T cell lymphoma and is in clinical trials for solid tumours as adjuvant therapy. One molecular target of vorinostat is INhibitor of Growth 2 (ING2), that together with ING1 serve as targeting subunits of the Sin3a HDAC complex. Treatment with sublethal (LD25-LD50) levels of vorinostat promoted breast cancer cell migration several-fold, which increased further upon ING1 knockout. These observations indicate that correct targeting of the Sin3a HDAC complex, and HDAC activity in general decreases luminal and basal breast cancer cell motility, suggesting that use of HDAC inhibitors as adjuvant therapies in breast cancers that are prone to metastasize may not be optimal and requires further investigation.


Asunto(s)
Neoplasias de la Mama , Inhibidores de Histona Desacetilasas , Femenino , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Cromatina , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Vorinostat/farmacología
2.
Cancers (Basel) ; 14(13)2022 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-35804879

RESUMEN

The Inhibitor of Growth (ING) proteins are a group of tumor suppressors with five conserved genes. A common motif of ING factors is the conserved plant homeodomain (PHD), with which they bind to chromatin as readers of the histone mark trimethylated histone H3 (H3K4me3). These genes often produce several protein products through alternative splicing events. Interestingly, ING1 and ING2 participate in the establishment of the repressive mSIN3a-HDAC complexes, whereas ING3, ING4, and ING5 are associated with the activating HAT protein complexes. In addition to the modulation of chromatin's structure, they regulate cell cycle transition, cellular senescence, repair of DNA damage, apoptosis, and angiogenic pathways. They also have fundamental effects on regulating cellular senescence in cancer cells. In the current review, we explain their role in cellular senescence based on the evidence obtained from cell line and animal studies, particularly in the context of cancer.

3.
Open Biol ; 11(9): 210077, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34493070

RESUMEN

Approximately 90% of cancer-related deaths can be attributed to a tumour's ability to spread. We have identified CG7379, the fly orthologue of human ING1, as a potent invasion suppressor. ING1 is a type II tumour suppressor with well-established roles in the transcriptional regulation of genes that control cell proliferation, response to DNA damage, oncogene-induced senescence and apoptosis. Recent work suggests a possible role for ING1 in cancer cell invasion and metastasis, but the molecular mechanism underlying this observation is lacking. Our results show that reduced expression of CG7379 promotes invasion in vivo in Drosophila, reduces the junctional localization of several adherens and septate junction components, and severely disrupts cell-cell junction architecture. Similarly, ING1 knockdown significantly enhances invasion in vitro and disrupts E-cadherin distribution at cell-cell junctions. A transcriptome analysis reveals that loss of ING1 affects the expression of several junctional and cytoskeletal modulators, confirming ING1 as an invasion suppressor and a key regulator of cell-cell junction integrity.


Asunto(s)
Neoplasias de la Mama/prevención & control , Comunicación Celular , Proteínas de Drosophila/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína Inhibidora del Crecimiento 1/metabolismo , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Humanos , Proteína Inhibidora del Crecimiento 1/genética , Células MCF-7 , Invasividad Neoplásica , Transcriptoma
4.
Cancers (Basel) ; 13(16)2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34439179

RESUMEN

The human telomerase is a key factor during tumorigenesis in prostate cancer (PCa). The androgen receptor (AR) is a key drug target controlling PCa growth and regulates hTERT expression, but is described to either inhibit or to activate. Here, we reveal that androgens repress and activate hTERT expression in a concentration-dependent manner. Physiological low androgen levels activate, while, notably, supraphysiological androgen levels (SAL), used in bipolar androgen therapy (BAT), repress hTERT expression. We confirmed the SAL-mediated gene repression of hTERT in PCa cell lines, native human PCa samples derived from patients treated ex vivo, as well as in cancer spheroids derived from androgen-dependent or castration resistant PCa (CRPC) cells. Interestingly, chromatin immuno-precipitation (ChIP) combined with functional assays revealed a positive (pARE) and a negative androgen response element (nARE). The nARE was narrowed down to 63 bp in the hTERT core promoter region. AR and tumor suppressors, inhibitor of growth 1 and 2 (ING1 and ING2, respectively), are androgen-dependently recruited. Mechanistically, knockdown indicates that ING1 and ING2 mediate AR-regulated transrepression. Thus, our data suggest an oppositional, biphasic function of AR to control the hTERT expression, while the inhibition of hTERT by androgens is mediated by the AR co-repressors ING1 and ING2.

5.
Ageing Res Rev ; 68: 101332, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33753287

RESUMEN

Cellular senescence occurs in response to diverse stresses (e.g., telomere shortening, DNA damage, oxidative stress, oncogene activation). A growing body of evidence indicates that alterations in multiple components of endocytic pathways contribute to cellular senescence. Clathrin-mediated endocytosis (CME) and caveolae-mediated endocytosis (CavME) represent major types of endocytosis that are implicated in senescence. More recent research has also identified a chromatin modifier and tumor suppressor that contributes to the induction of senescence via altered endocytosis. Here, molecular regulators of aberrant endocytosis-induced senescence are reviewed and discussed in the context of their capacity to serve as senescence-inducing stressors or modifiers.


Asunto(s)
Senescencia Celular , Endocitosis , Daño del ADN , Humanos , Estrés Oxidativo
6.
Cancer Cell Int ; 20: 398, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32831651

RESUMEN

BACKGROUND: Oral squamous cell carcinoma (OSCC), a form of head and neck squamous cell carcinoma (HNSCC) has a poor 5-year survival rate. OSCC patients are often treated with cisplatin but resistance to chemotherapy is often observed. This makes it important identification of alternative therapeutic targets which will result in more favorable outcome in OSCC patients. The plant homeodomain (PHD)-containing protein Inhibitor of Growth family of tumor suppressor proteins (p33ING1b) has been indicated as a tumor suppressor in different cancers including OSCC. This protein has been shown to function by modulating transcriptional activity of p53; however, the exact mechanism(s) are not well defined. METHODS: Expression of total and acetylated p53 and p33ING1b protein was determined in OSCC cell lines YD-9, YD-8, and YD-38 by immunoblot analysis. Effect of modulation of p33ING1b protein expression on acetylation of p53 and cell proliferation was determined by immunoblot and MTT assay. Effect of modulation of p33ING1b protein expression on transactivation of p53 was assessed by heterologous promoter-based reporter and chromatin immunoprecipitation. Effect of modulation of expression of p33ING1b on SIR2 mRNA and protein was determined by quantitative real-time PCR and immunoblot analyses. Impact of modulation of p33ING1b alone or in combination with SIR2 on chemosensitivity of YD-9 and YD-8 cells to cisplatin was determined in time and dose-dependent cell proliferation assays. RESULTS: Here, using a panel of OSCC cell lines with wild type or mutant p53, we show that p33ING1b expression is correlated to acetylation of p53 at lysine 382 residue. Increased acetylation of p53 following overexpression of p33ING1b was associated with increased expression of the pro-apoptotic proteins BAX, p21, and cleaved-Caspase 3, and decreased cell proliferation. Reporter assays with p21 and BAX promoters showed that p33ING1b expression levels directly correlated to promoter activity of these 2 genes. Chromatin immunoprecipitation assay showed that transcriptional regulation of p21 and BAX by acetylated p53 is dependent on expression level of p33ING1b. Differential acetylation of p53 following modulation of p33ING1b expression was indirect. Expression of p33ING1b was found to be inversely correlated to the NAD-dependent deacetylase silent information regulator 2 (SIR2). SIR2 was transcriptionally regulated by p33ING1b. Relative expression of p33ING1b was found to dictate chemosensitivity of OSCC cell lines to cisplatin treatment. Concomitant overexpression of p33ING1b and knockdown of SIR2 had a synergistic effect on chemosensitivity of OSCC cell lines to cisplatin, compared to either overexpression of p33ING1b or knockdown of SIR2 alone. CONCLUSIONS: The results from the current study thus elucidate that p33ING1b regulates p53 acetylation irrespective of p53 mutation and subsequent transactivation by transcriptional regulation of SIR2 expression. The results also indicate that p33ING1b and SIR2 are potentially attractive therapeutic targets.

7.
Transl Cancer Res ; 9(5): 3623-3633, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-35117725

RESUMEN

BACKGROUND: Inhibitor growth protein 1 (ING1) is a tumor suppressor, and its down-regulation is involved in the progression and aggressive phenotypes of human malignancies through its interactions with the H3K4me3 and p53. METHODS: We collected datasets to analyze the relationship between ING1b mRNA expression and accumulative survival rate, and carried out immunohistochemistry analyses to determine the expression profiles of the p33ING1 protein on the mouse, normal human, and human cancer tissue microarrays. RESULTS: Compared with normal tissues, the ING1b mRNA was highly expressed in various types of cancer tissues, including, colorectal, lung, and breast cancers, and was positively correlated with the overall survival rate of gastric cancer patients. In mouse tissues, the subcellular location of p33ING1 was frequently nuclear; however, it was occasionally cytoplasmic or nucleocytoplasmic. There was a positive detection in the neuron body, a part of glial cells, the glandular epithelium of the stomach, intestines, breast, hepatocytes, heart, skeletal muscle cells, the bronchial and alveolar epithelium, and nephric tubules. In human tissues, the p33ING1 protein, apart from its cytoplasmic distribution, was distributed in the nuclei of the tongue, esophagus, stomach, intestine, lung, trachea, skin, appendix, cervix, endometrium, ovary, and breast. p33ING1 immunoreactivity was strongly detected in the stomach, trachea, skin, cervix, and breast, while it was weak in the other tissues. The positive rate of p33ING1 was 41.0% in the tested cancer entities (489/1,194). In general, p33ING1 expression was restricted to only the cytoplasm for all cancers, whereas it was found in the nucleus of renal clear cells, ovarian and colorectal cancers. Among them, p33ING1 was expressed in more than half of squamous cell carcinomas derived from the esophagus and cervix, while it was rarely expressed in hepatocellular (21.0%) and renal clear cell carcinoma (19.4%). CONCLUSIONS: The findings suggest that p33ING1 might be participated in the repair and regeneration of organs or tissues the repair and regeneration of organs or tissue, and the carcinogenesis of the highly proliferative epithelium.

8.
Cancers (Basel) ; 11(10)2019 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-31640185

RESUMEN

Non-small cell lung cancer (NSCLC) has been the leading cause of cancer-related death worldwide, over the last few decades. Survival remains extremely poor in the metastatic setting and, consequently, innovative therapeutic strategies are urgently needed. Inhibitor of Growth Gene 2 (ING2) is a core component of the mSin3A/Histone deacetylases complex (HDAC), which controls the chromatin acetylation status and modulates gene transcription. This gene has been characterized as a tumor suppressor gene and its status in cancer has been scarcely explored. In this review, we focused on ING2 and other mSin3A/HDAC member statuses in NSCLC. Taking advantage of existing public databases and known pharmacological properties of HDAC inhibitors, finally, we proposed a therapeutic model based on an ING2 biomarker-guided strategy.

9.
Mech Ageing Dev ; 177: 109-117, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29928906

RESUMEN

Replicative capacity of normal human cells decreases as telomeric sequence is lost at each division. It is believed that when a subset of chromosomes reach a critically short length, an ATM-initiated and p53-mediated transcriptional response inhibits cell growth, promoting cell senescence. In addition to loss of telomeric sequence, senescence can be induced by other stresses including ionizing radiation, oxidative damage, chemical crosslinkers like the chemotherapeutic agent cisplatin, as well as overactivation of oncogenes and tumor suppressors. Our group found that the expression of an isoform of the INhibitor of Growth 1 gene called ING1a increases approximately 10-fold as fibroblasts approach senescence and that forced expression rapidly induces a senescent phenotype in primary diploid fibroblasts, epithelial and endothelial cells that resembles replicative senescence by most physical and biochemical measures. ING1a induces these changes through strongly inhibiting endocytosis to block mitogen signaling by inducing the expression of intersectin 2, a key scaffolding protein of the endosomal pathway. This, in turn increases the expression of Rb and of p57Kip2 and p16INK4a that serve to maintain Rb is an active, growth inhibitory state. The ING1a model is currently being used to better understand the mechanism(s) responsible for activating Rb to enforce the senescent state.


Asunto(s)
Senescencia Celular/fisiología , Regulación de la Expresión Génica/fisiología , Proteína Inhibidora del Crecimiento 1/biosíntesis , Modelos Biológicos , Proteínas Adaptadoras del Transporte Vesicular/biosíntesis , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Endocitosis/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/patología , Humanos , Proteína Inhibidora del Crecimiento 1/genética , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
10.
Biomed Pharmacother ; 108: 1628-1639, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30372865

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) have been consistently demonstrated to be involved in non-small cell lung cancer (NSCLC) as either tumor oncogenes or tumor suppressors. However, the detailed role of miR-500 and miR-628 in NSCLC remain poorly understood. METHODS: The expressions of miR-500 and miR-628 in NSCLC tissues and cell lines were measured by quantitative real-time PCR (qRT-PCR). Cells migration, invasion, proliferation, adhesion and apoptosis abilities were test to analyze the biological functions of miR-500 and miR-628 in NSCLC. A bioinformatic analysis was conducted to predict the target genes regulated by miR-500 and miR-628 using TargetScan (http://www.targetscan.org/mamm/). Luciferase reporter assay was employed to validate the direct targeting of ING1 by miR-500 and miR-628. RESULTS: In this study, miR-500 and miR-628 were up-regulated with NSCLC tissues. Furthermore, inhibition of miR-500 and miR-628 significantly suppressed NSCLC cells proliferation, migration, invasion and adhesion, and induced NSCLC cells apoptosis. Additionally, the result showed that ING1 functioned as the direct target for miR-500 and miR-628, which was a core tumor suppressor in regulating NSCLC progression. Over-expression of ING1 could dramatically inhibit NSCLC cells proliferation, migration and invasion, and promote cells apoptosis. CONCLUSION: These results brought new insights into the oncogenic role of miR-500 and miR-628 in NSCLC, indicating that miR-500 and miR-628 might be the novel biomarkers for the diagnosis and prognosis of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Movimiento Celular/fisiología , Proteína Inhibidora del Crecimiento 1/biosíntesis , Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/prevención & control , Proliferación Celular/fisiología , Regulación hacia Abajo/fisiología , Marcación de Gen/métodos , Humanos , Proteína Inhibidora del Crecimiento 1/antagonistas & inhibidores , Neoplasias Pulmonares/prevención & control , MicroARNs/antagonistas & inhibidores
11.
Cell Mol Biol (Noisy-le-grand) ; 64(5): 66-72, 2018 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-29729696

RESUMEN

The effect of deregulation of nuclear export mediated by exportin-1, with consequent cellular mislocalization of p33ING1b, a member of the tumor suppressor gene family, has not been previously investigated in head and neck squamous cell cancer (HNSCC). We evaluated the effect of reversing cytoplasmic p33ING1b localization through inhibition of exportin-1 by leptomycin B (LMB) and the effect of nuclear entrapment of p33ING1b on molecular alterations in primary and metastatic HNSCC lines. The expression and location of exportin-1 and p33ING1b were analyzed by a quantitative real­time reverse transcription polymerase chain reaction PCR (qRT-PCR), a Western blot, and immunostaining. Cell proliferation and migration assays were conducted to determine the effect of exportin-1 inhibition on the cell lines. Exportin-1 was overexpressed in metastatic HNSCC, whereas p33ING1b was poorly expressed. Exportin-1 inhibition induced nuclear entrapment and upregulation of p33ING1b, extensive apoptosis, and growth arrest. It also suppressed cell migration. Cytoplasmic p33ING1b-mediated regulation of cell growth and nuclear entrapment of p33ING1b via inhibition of exportin-1 may be a key mechanism for inducing HNSCC apoptosis.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Proteínas de Homeodominio/genética , Carioferinas/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Supresoras de Tumor/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Ácidos Grasos Insaturados/farmacología , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Proteínas de Homeodominio/metabolismo , Humanos , Carioferinas/antagonistas & inhibidores , Carioferinas/metabolismo , Metástasis de la Neoplasia , Cultivo Primario de Células , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Proteína Exportina 1
12.
Neuroscience ; 369: 248-260, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29158107

RESUMEN

Epigenetic regulation of activity-induced gene expression involves multiple levels of molecular interaction, including histone and DNA modifications, as well as mechanisms of DNA repair. Here we demonstrate that the genome-wide deposition of inhibitor of growth family member 1 (ING1), which is a central epigenetic regulatory protein, is dynamically regulated in response to activity in primary cortical neurons. ING1 knockdown leads to decreased expression of genes related to synaptic plasticity, including the regulatory subunit of calcineurin, Ppp3r1. In addition, ING1 binding at a site upstream of the transcription start site (TSS) of Ppp3r1 depends on yet another group of neuroepigenetic regulatory proteins, the Piwi-like family, which are also involved in DNA repair. These findings provide new insight into a novel mode of activity-induced gene expression, which involves the interaction between different epigenetic regulatory mechanisms traditionally associated with gene repression and DNA repair.


Asunto(s)
Corteza Cerebral/metabolismo , Epigénesis Genética , Proteína Inhibidora del Crecimiento 1/metabolismo , Neuronas/metabolismo , Animales , Proteínas Argonautas/metabolismo , Proteínas de Unión al Calcio , Células Cultivadas , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteína Inhibidora del Crecimiento 1/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos C57BL , Proteínas Musculares/metabolismo , ARN Mensajero/metabolismo
13.
J Mol Med (Berl) ; 94(10): 1167-1179, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27305909

RESUMEN

The androgen receptor (AR) is a transcriptional factor that has a pivotal role in the development of normal and also cancerous prostate. Therefore, analyzing AR signaling is essential to understand cancerogensis and proliferation of prostate cancer (PCa). Inhibitor of growth 1 (ING1) and ING2 are tumor suppressors with reduced expression in many cancer types. There are also indications of misregulation of ING1 and ING2 in PCa. However, the roles of ING1 and ING2 in PCa and AR signaling are poorly understood. Here, we show that surprisingly the ING1b knockdown (KD) represses AR-mediated transactivation on AR key target genes in the human LNCaP PCa cells. This is associated with growth reduction of LNCaP cells by ING1 KD. In line with this, using Ing1 knockout (KO) mice, we provide further evidence that ING1 deficiency downregulates prostate-specific AR target genes in vivo. Further analyses suggest that KD of ING1b results in induction of both cellular senescence and the cell cycle inhibitor p16 INK4a . The unexpected finding that the ING1 KD results in growth inhibition was further analyzed and can be explained by a compensatory mechanism through enhanced levels of ING2 protein in ING1-deficient condition. Accordingly, the data suggest that ING2 interacts with AR and hampers the AR transcriptional activation, causes growth arrest, and induces cellular senescence. The data further suggest that ING2 upregulates p16 INK4a , which is a novel target for ING2. Taken together, our data suggest that ING2 is a novel corepressor for AR. ING2 levels are increased upon downregulation of ING1 expression indicating a compensatory mechanism and suggests a novel crosstalk between ING1 and ING2 tumor suppressors to inhibit AR signaling and induce cellular senescence in PCa cells. KEY MESSAGE: • The tumor suppressors ING1 and 2 are dysregulated in human prostate cancer. • ING1 deficiency reduces AR-mediated gene expression in vitro and in vivo. • ING2, like ING1, inhibits AR-mediated transactivation and prostate cancer cell growth. • ING1 regulates ING2. • ING1 and ING2 crosstalk with each other to inhibit AR signaling in prostate cancer.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Receptores Androgénicos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Senescencia Celular , Humanos , Proteína Inhibidora del Crecimiento 1 , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones Noqueados , Proteínas Nucleares/genética , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Vesículas Seminales/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética
14.
J Mol Cell Biol ; 8(3): 207-20, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26993046

RESUMEN

The androgen receptor (AR) signaling is critical for prostate cancer (PCa) progression to the castration-resistant stage with poor clinical outcome. Altered function of AR-interacting factors may contribute to castration-resistant PCa (CRPCa). Inhibitor of growth 1 (ING1) is a tumor suppressor that regulates various cellular processes including cell proliferation. Interestingly, ING1 expression is upregulated in senescent primary human prostate cells; however, its role in AR signaling in PCa was unknown. Using a proteomic approach by surface-enhanced laser desorption ionization-mass spectrometry (SELDI-MS) combined with immunological techniques, we provide here evidence that ING1b interacts in vivo with the AR. The interaction was confirmed by co-immunoprecipitation, in vitro GST-pull-down, and quantitative intracellular colocalization analyses. Functionally, ING1b inhibits AR-responsive promoters and endogenous key AR target genes in the human PCa LNCaP cells. Conversely, ING1b knockout (KO) mouse embryonic fibroblasts (MEFs) exhibit enhanced AR activity, suggesting that the interaction with ING1b represses the AR-mediated transcription. Also, data suggest that ING1b expression is downregulated in CRPCa cells compared with androgen-dependent LNCaP cells. Interestingly, its ectopic expression induces cellular senescence and reduces cell migration in both androgen-dependent and CRPCa cells. Intriguingly, ING1b can also inhibit androgen-induced growth in LNCaP cells in a similar manner as AR antagonists. Moreover, ING1b upregulates different cell cycle inhibitors including p27(KIP1), which is a novel target for ING1b. Taken together, our findings reveal a novel corepressor function of ING1b on various AR functions, thereby inhibiting PCa cell growth.


Asunto(s)
Senescencia Celular , Proteínas Co-Represoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Proteína Inhibidora del Crecimiento 1 , Masculino , Ratones , Células 3T3 NIH , Neoplasias de la Próstata/genética , Unión Proteica , Transcripción Genética , Activación Transcripcional/genética
15.
Oncol Lett ; 10(3): 1517-1522, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26622701

RESUMEN

Inhibitor of growth 1b (ING1b) is considered to be a class II tumor suppressor gene. Although decreased expression of p33ING1b has previously been reported in colorectal cancer (CRC), its role in CRC has remained to be elucidated. The present study was designed to assess the function of p33ING1b in CRC and to further evaluate its underlying mechanisms of action. Western blot analysis confirmed that ING1b gene expression was significantly decreased in CRC tissues compared with that of adjacent non-tumorous colorectal tissues. Furthermore, recombinant adenovirus-mediated ectopic expression of p33ING1b resulted in growth inhibition, G1-phase cell cycle arrest and apoptosis in the SW480, HT29 and LoVo colorectal adenocarcinoma cell lines. The results suggested that the downregulation of ING1b contributes to colorectal carcinogenesis and that ectopic expression of ING1b may be a potentially useful therapeutic approach for CRC.

16.
Oncotarget ; 6(33): 34118-27, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26439691

RESUMEN

Cell senescence contributes to organismal aging and is induced by telomere erosion and an ensuing DNA damage signal as cells reach the end of their replicative lifespan in vitro or in vivo. Stresses induced by oncogene or tumor suppressor hyperactivation, oxidative stress, ionizing radiation and other DNA damaging agents result in forms of stress induced premature senescence (SIPS) that show similarities to replicative senescence. Since replicative senescence and SIPS occur over many days and many population doublings of the mass cultures of primary cells used to study senescence, the sequence of events that occur downstream of senescence signaling can be challenging to define. Here we compare a new model of ING1a-induced senescence with several other forms of senescence. The ING1a epigenetic regulator synchronously induces senescence in mass cultures several-fold faster than all other agents, taking 24 and 36 hours to activate the Rb/ p16INK4a, but not the p53 tumor suppressor axis to efficiently induce senescence. ING1a induces expression of intersectin 2, a scaffold protein necessary for endocytosis, altering the stoichiometry of endocytosis proteins, subsequently blocking growth factor uptake leading to activation of Rb signaling to block cell growth. ING1a acts as a novel link in the activation of the Rb pathway that can impose senescence in the absence of activating p53-mediated DNA damage signaling, and should prove useful in defining the molecular events contributing to Rb-induced senescence.


Asunto(s)
Envejecimiento Prematuro/genética , Senescencia Celular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Proteína de Retinoblastoma/metabolismo , Estrés Fisiológico/genética , Proteínas Supresoras de Tumor/genética , Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/biosíntesis , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN/genética , Endocitosis/fisiología , Células Endoteliales/metabolismo , Humanos , Proteína Inhibidora del Crecimiento 1 , Queratinocitos/metabolismo , Homeostasis del Telómero/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
Practical Oncology Journal ; (6): 254-258, 2015.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-499399

RESUMEN

Tumor inhibiting protein p33ING1b is the key expressive product of inhibitor of growth 1.It plays critical role in cell multiplication ,period control ,senescence ,repair of DNA damage ,apoptosis ,and chroma-tin remodeling.The abnormal expression of p33ING1b is closely related to the occurrence and development of tumor.This paper reviews tumor inhibiting protein p 33ING1b in the research development of tumors in digestive system.

18.
Oncol Lett ; 7(5): 1639-1644, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24765192

RESUMEN

The present study aimed to investigate the feasibility of detecting p33 inhibitor of growth 1b (p33ING1b) gene methylation in fecal DNA as a screening method for colorectal carcinoma (CRC) and precancerous lesions. The methylation of p33ING1b was analyzed in fecal samples from 61 patients with CRCs, 27 patients with precancerous lesions (advanced adenoma) and 20 normal individuals by nested methylation-specific polymerase chain reaction (nMSP) and fecal occult blood test. Methylated p33ING1b was detected in 73.77% of CRC patients and 62.96% of adenoma patients. By contrast, only 5% of normal individuals had methylated p33ING1b. These results indicated 73.77% sensitivity for detecting CRC, 62.96% sensitivity for detecting precancerous lesions and 95% specificity of the assay for detecting CRCs and precancerous lesions. The detection of p33ING1b methylation status by incubation of DNA contained in agarose beads for bisulfite modification, followed by nMSP, is a promising non-invasive screening method for CRCs and precancerous lesions.

19.
Cancer Lett ; 345(1): 1-16, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24333729

RESUMEN

ING genes (ING1-5) were identified has tumor suppressor genes. ING proteins are characterized as Type II TSGs since they are involved in the control of cell proliferation, apoptosis and senescence. They may also function as Type I TSGs since they are also involved in DNA replication and repair. Most studies have reported that they are frequently lost in human tumors and epigenetic mechanisms or misregulation of their transcription may be involved. Recently, studies have described that this loss may be caused by microRNA inhibition. Here, we summarize the current knowledge on ING functions, their involvement in tumor suppression and, in order to give a full assessment of the current knowledge, we review all the studies that have examined ING status in human cancers.


Asunto(s)
Genes Supresores de Tumor , Péptidos y Proteínas de Señalización Intracelular/genética , MicroARNs/genética , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Animales , Humanos , Proteína Inhibidora del Crecimiento 1 , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo
20.
The Journal of Practical Medicine ; (24): 1391-1394, 2014.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-451345

RESUMEN

Objective To discuss the expressions and clinical significance of p53 and p33ING1b in human psoriasis and basal cell carcinoma (BCC). Methods Immunohistochemistry EnVision technique was used to detect the expressions of p53 and p33ING1b in samples of 36 psoriasis vulgaris, 28 BCC and 14 normal skins. Results The expression of p53 increased while p33ING1b had a degressive expression in the control group, the psoriasis group and the BCC group. It was found significant statistical difference between the two groups (all P < 0.05). Prominent positive correlation between p53 and p33ING1b were found in both psoriasis group and BCC group (all P<0.05). Conclusions p53 coacts with p33ING1b at local lesions of abnormal proliferative diseases . It′s one of the most prominent mechanisms contributing to deviant cell proliferation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA