Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
BMC Biotechnol ; 21(1): 42, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34281556

RESUMEN

BACKGROUND: Protein C receptor (Procr) has recently been shown to mark resident adult stem cells in the mammary gland, vascular system, and pancreatic islets. More so, high Procr expression was also detected and used as indicator for subsets of triple-negative breast cancers (TNBCs). Previous study has revealed Procr as a target of Wnt/ß-catenin signaling; however, direct upstream regulatory mechanism of Procr remains unknown. To comprehend the molecular role of Procr during physiology and pathology, elucidating the upstream effectors of Procr is necessary. Here, we provide a system for screening negative regulators of Procr, which could be adapted for broad molecular analysis on membrane proteins. RESULTS: We established a screening system which combines CRISPR-Cas9 guided gene disruption with fluorescence activated cell sorting technique (FACS). CommaDß (murine epithelial cells line) was used for the initial Procr upstream effector screening using lentiviral CRISPR-gRNA library. Shortlisted genes were further validated through individual lentiviral gRNA infection followed by Procr expression evaluation. Adam17 was identified as a specific negative inhibitor of Procr expression. In addition, MDA-MB-231 cells and Hs578T cells (human breast cancer cell lines) were used to verify the conserved regulation of ADAM17 over PROCR expression. CONCLUSION: We established an efficient CRISPR-Cas9/FACS screening system, which identifies the regulators of membrane proteins. Through this system, we identified Adam17 as the negative regulator of Procr membrane expression both in mammary epithelial cells and breast cancer cells.


Asunto(s)
Proteína ADAM17/metabolismo , Receptor de Proteína C Endotelial/genética , Lentivirus/genética , Glándulas Mamarias Humanas/enzimología , Proteína ADAM17/genética , Secuencia de Bases , Línea Celular , Regulación hacia Abajo , Receptor de Proteína C Endotelial/metabolismo , Biblioteca de Genes , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Lentivirus/metabolismo , ARN Guía de Kinetoplastida/genética
2.
Elife ; 102021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34096503

RESUMEN

RAS-like (RAL) GTPases function in Wnt signalling-dependent intestinal stem cell proliferation and regeneration. Whether RAL proteins work as canonical RAS effectors in the intestine and the mechanisms of how they contribute to tumourigenesis remain unclear. Here, we show that RAL GTPases are necessary and sufficient to activate EGFR/MAPK signalling in the intestine, via induction of EGFR internalisation. Knocking down Drosophila RalA from intestinal stem and progenitor cells leads to increased levels of plasma membrane-associated EGFR and decreased MAPK pathway activation. Importantly, in addition to influencing stem cell proliferation during damage-induced intestinal regeneration, this role of RAL GTPases impacts on EGFR-dependent tumourigenic growth in the intestine and in human mammary epithelium. However, the effect of oncogenic RAS in the intestine is independent from RAL function. Altogether, our results reveal previously unrecognised cellular and molecular contexts where RAL GTPases become essential mediators of adult tissue homeostasis and malignant transformation.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Receptores ErbB/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Receptores de Péptidos de Invertebrados/metabolismo , Células Madre/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Animales , Animales Modificados Genéticamente , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Endocitosis , Receptores ErbB/genética , Femenino , Humanos , Hiperplasia , Mucosa Intestinal/patología , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Glándulas Mamarias Humanas/enzimología , Glándulas Mamarias Humanas/patología , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Receptores de Péptidos de Invertebrados/genética , Transducción de Señal , Células Madre/patología , Proteínas de Unión al GTP ral/genética
3.
Int Immunopharmacol ; 91: 107324, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33385711

RESUMEN

It is well-established that lysine-specific demethylase 1 (LSD1) is the first identified histone demethylase. Based on its demethylase enzymatic activity, LSD1 plays a pivotal role in vast range of cellular processes and cancers, but the understanding of its effects on inflammation is relatively limited. Using in vivo models of lipopolysaccharide (LPS)-induced inflammation and in vitro assays in mouse mammary epithelial cells, we identified the novel regulatory roles and underlying mechanisms of LSD1 on LPS-induced mastitis. Mammary gland and cells were collected for the following experiments after treatment. Histological changes were determined by H&E. Western blot analysis was used to detect the protein expression. ELISA and real-time PCR were used to evaluate protein and mRNA expression of inflammatory genes. Our results showed that LPS treatment resulted in a significant increase in LSD1 protein expression. GSK-LSD1 is a selective inhibitor of LSD1 enzyme activity. Treatment of mice with GSK-LSD1 inhibited LSD1 activity, reduced inflammatory cells recruitment to tissues and attenuated LPS-induced damage in mammary gland. Mechanistic investigations suggested that LSD1 inhibition led to the increase of histone H3K4me2 and H3K9me2. Furthermore, GSK-LSD1 inhibition of LSD1 further inhibited nuclear factor κ-B (NF-κB) signaling cascades, and subsequently inhibited the production of cytokines (TNF-α, IL-6 and IL-1ß) in mammary gland. Taken together, our data reveal LSD1 as a potential regulator of inflammation and improve our understanding of epigenetic control on inflammation.


Asunto(s)
Epigénesis Genética , Células Epiteliales/enzimología , Histona Demetilasas/metabolismo , Glándulas Mamarias Humanas/enzimología , Mastitis/enzimología , Animales , Antiinflamatorios/farmacología , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/genética , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/patología , Mastitis/inducido químicamente , Mastitis/genética , Mastitis/prevención & control , Ratones Endogámicos BALB C , FN-kappa B/metabolismo
4.
Toxicol Mech Methods ; 29(9): 686-692, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31364906

RESUMEN

Life-long estrogen exposure is one of the major risk factors in the development and progression of breast cancer. However, little is known about the molecular mechanisms, by which chronic exposure to estrogen contributes to breast carcinogenesis. The aim of the present study was to investigate the effects of long-term exposure with 4-hydroxyestradiol (4-OHE2) on acquired cancer characteristics of human mammary epithelial MCF-10A cells. The possible regulators were further studied in chronic 4-OHE2-treated MCF-10A cells. We observed that MCF-10A cells long-term exposed to 4-OHE2 acquire the characteristics of cancer cells, such as enhanced cell growth, EMT properties, and increased migration and invasiveness. Moreover, the expression of CYP1B1 was significantly elevated in long-term 4-OHE2-treated MCF-10A cells. Block of CYP1B1 significantly reduced the cancer cell characteristics in long-term 4-OHE2-treated MCF-10A cells. Our results indicated that 4-OHE2 mediated enhanced cancer cell characteristics in mammary epithelial cells are an important key event for breast carcinogenesis process. CYP1B1 partially contributes to the 4-OHE2 induced cancer cell characteristics in MCF-10A cells. Targeting CYP1B1 might offer a new strategy for the treatment of estrogen-induced breast cancer.


Asunto(s)
Carcinogénesis/inducido químicamente , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP1B1/biosíntesis , Transición Epitelial-Mesenquimal/efectos de los fármacos , Estrógenos de Catecol/toxicidad , Carcinogénesis/metabolismo , Carcinogénesis/patología , Técnicas de Cultivo de Célula , Línea Celular , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/patología , Femenino , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/enzimología , Regulación hacia Arriba
5.
Methods Mol Biol ; 1952: 193-199, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30825175

RESUMEN

To explore the physiological or pathological roles of proteases, it is important to be able to detect and precisely localize them in a tissue, to differentiate between inactive and active forms, as well as to quantify and determine the nature of the enzyme that degrades a given substrate. Here we present an in situ gelatin zymography method that allows for a precise localization of active gelatin-degrading enzymes in a tissue section. In this method, dye-quenched gelatin is put on top of a tissue section. During an incubation period, active gelatinolytic enzymes will degrade the substrate and fluorescent signals are emitted from the locations of these enzymes.


Asunto(s)
Pruebas de Enzimas/métodos , Gelatinasas/metabolismo , Microscopía Fluorescente/métodos , Microtomía/métodos , Animales , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/metabolismo , Gelatina/análisis , Gelatina/metabolismo , Gelatinasas/análisis , Humanos , Glándulas Mamarias Humanas/química , Glándulas Mamarias Humanas/enzimología , Glándulas Mamarias Humanas/ultraestructura , Ratones , Especificidad por Sustrato , Adhesión del Tejido/métodos , Fijación del Tejido/métodos
6.
Int J Cancer ; 145(3): 817-829, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30671946

RESUMEN

The hotspot mutation H1047R in the oncogenic PIK3CA gene is frequently detected in breast cancer and enhances the enzymatic activity of PI3K to activate AKT/mTOR signaling cascade. Aberrant elevated PI3K activation has been reported to promote the tumorigenesis of breast cancer, but the mechanisms underlying are still needed to be elucidated. Here, we found that continuously activating PIK3CAH1047R conferred human mammary epithelial MCF-10A cells to cellular senescence upon serum-starvation. Similarly, breast cancer T47D and HCC1954 cells harboring H1047R mutation were senescent when cells were deprived of serum. PI3K/AKT/mTOR axis but not p53 or RB might be required for the induction of senescence. Notably, membrane metallo-endopeptidase (MME) was identified as a downstream effector of PI3K to mediate the induction of senescence, which might be associated with its glycosylation. Senescent cells elicited a distinct secretome dependent on PI3K and MME. Specifically, IL-6 promoted the proliferation of normal cells and CCL2 induced the M2-like polarization of macrophages, which might create an immunosuppressive microenvironment during the initiation and/or development of breast cancer. This study shed new light on the tumorigenesis induced by hyper-activated PI3K and might provide new clues for the prevention and therapy of breast cancer.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Metaloendopeptidasas/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Senescencia Celular/fisiología , Quimiocina CCL2/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Células Epiteliales/citología , Células Epiteliales/enzimología , Glicosilación , Humanos , Interleucina-6/metabolismo , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/enzimología , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
7.
Anim Sci J ; 90(2): 214-221, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30556368

RESUMEN

The aim of this study was to determine the effects of isonitrogenous and isocaloric diets containing different qualities of forages and concentrate content on milk fat composition and genes that encode mammary lipogenic enzymes in dairy cows. A total of 20 Holstein cows were assigned to 1 of 2 treatment diets composed of either mixed forages (MF, starch : 21.50%) or corn stover forage (CS, starch : 25.39%). Mammary tissue biopsies were performed to analyze the mRNA expression of lipogenic enzymes. Dry matter intake, body weight, milk protein, and lactose were not affected by treatments. The milk yield, fat content and saturated fatty acid (SFA) and short- and medium-chain fatty acid (SMFA) contents in milk were lower in the CS diet than in the MF diet, but the unsaturated FA and long-chain FA contents were higher. Genes involved in de novo FA synthesis, FA uptake and transport, and Δ9-desaturation were lower in the CS treatment than in the MF treatment. No effects on the nuclear transcription factors were observed between the two treatments. The data indicated that corn stover diet reduced the milk yield, fat content, SMFA, and SFA contents in milk, as well as the gene expression of mammary lipogenic enzymes in dairy cows.


Asunto(s)
Alimentación Animal , Bovinos/metabolismo , Bovinos/fisiología , Dieta/veterinaria , Ácidos Grasos/metabolismo , Lactancia , Lipogénesis/genética , Glándulas Mamarias Humanas/enzimología , Glándulas Mamarias Humanas/metabolismo , Leche/metabolismo , Acetil-CoA Carboxilasa/genética , Acetil-CoA Carboxilasa/metabolismo , Alimentación Animal/análisis , Animales , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Proteína 3 de Unión a Ácidos Grasos/genética , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Ácidos Grasos/biosíntesis , Femenino , Expresión Génica , Humanos , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Proteínas de la Leche/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Almidón , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Zea mays
8.
Biochem Biophys Res Commun ; 505(2): 385-391, 2018 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-30262139

RESUMEN

In vitro, the rat Fatty Acid Desaturase 3 (FADS3) gene was shown to code for an enzyme able to catalyze the unexpected Δ13-desaturation of trans-vaccenic acid, producing the trans11,cis13-conjugated linoleic acid (CLA) isomer. FADS3 may therefore be the first methyl-end trans-vaccenate Δ13-desaturase functionally characterized in mammals, but the proof of this concept is so far lacking in vivo. The present study therefore aimed at investigating further the putative in vivo synthesis of trans11,cis13-CLA from dietary trans-vaccenic acid in rodents. During one week of pregnancy and two weeks post-partum, Sprague-Dawley female rats were fed two diets either high (10.0% of fatty acids and 3.8% of energy intake) or low (0.4% of fatty acids and 0.2% of energy intake) in trans-vaccenic acid. The trans11,cis13-CLA was specifically detected, formally identified and reproducibly quantified (0.06% of total fatty acids) in the mammary gland phospholipids of lactating female rats fed the high trans-vaccenic acid-enriched diet. This result was consistent with FADS3 mRNA expression being significantly higher in the lactating mammary gland than in the liver. Although the apparent metabolic conversion is low, this physiological evidence demonstrates the existence of this new pathway described in the lactating mammary gland and establishes the FADS3 enzyme as a reliable mammalian trans-vaccenate Δ13-desaturase in vivo.


Asunto(s)
Ácido Graso Desaturasas/metabolismo , Ácidos Linoleicos Conjugados/metabolismo , Glándulas Mamarias Humanas/metabolismo , Ácidos Oléicos/metabolismo , Animales , Catálisis , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Ácido Graso Desaturasas/genética , Femenino , Humanos , Lactancia , Ácidos Linoleicos Conjugados/biosíntesis , Glándulas Mamarias Humanas/enzimología , ARN Mensajero/metabolismo , Ratas
9.
PLoS One ; 13(9): e0203397, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30180179

RESUMEN

Vaccinia-related kinase 1 (VRK1) is a pro-proliferative nuclear kinase. Mice engrafted with VRK1-depleted MDA-MB-231 breast cancer cells have been shown to develop fewer distal metastases than controls, suggesting VRK1 might play a role in cell migration, invasion, and/or colonization. In work described herein, we investigated the impact of VRK1 overexpression on human mammary epithelial cells. In 2D culture, VRK1 overexpression diminishes cell migration and invasion and impairs the migration-associated processes of cell spreading and cytoskeletal rearrangement. VRK1-overexpressing cells show reduced accumulation of the mesenchymal marker vimentin and increased accumulation of the epithelial markers E-cadherin and claudin-1. VRK1 overexpression also leads to reduced levels of the transcriptional repressors snail, slug, and twist1. Cumulatively, these data indicate that VRK1 overexpression augments the epithelial properties of both MCF10a and MDA-MB-231 cells. We further studied the impact of VRK1 on the epithelial properties of MCF10a cells in 3D matrigel culture, in which cells proliferate and form epithelial sheets that mature into hollow spherical acini. VRK1 overexpression significantly accelerates the initial stages of cell proliferation, leading to larger acini that nevertheless differentiate and mature. Our analysis of human tumor tissue microarrays (TMAs) revealed that VRK1 protein levels are higher in lymph node metastases than in patient-matched mammary tumors. Using public databases, we determined that VRK1 is among the top 10% of overexpressed transcripts in multiple subtypes of invasive breast cancer, and that high levels of VRK1 expression are correlated with decreased relapse-free survival. In sum, overexpression of VRK1, by regulating the transcription repressors snail, slug, and twist1, can promote a mesenchymal-to-epithelial transition (MET) in cell culture. VRK1-mediated MET might facilitate the colonization of distal sites by metastatic breast cancer cells, providing some insight into the frequent association of VRK1 overexpression with breast malignancies and the correlation between VRK1 overexpression and poor clinical outcome.


Asunto(s)
Neoplasias de la Mama/enzimología , Movimiento Celular , Transición Epitelial-Mesenquimal , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Glándulas Mamarias Humanas/enzimología , Proteínas de Neoplasias/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Cadherinas/biosíntesis , Cadherinas/genética , Línea Celular Tumoral , Claudina-1/biosíntesis , Claudina-1/genética , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Metástasis Linfática , Glándulas Mamarias Humanas/patología , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética
10.
Cell Death Dis ; 9(3): 259, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449545

RESUMEN

Cellular senescence response is (i) activated by numerous stresses, (ii) is characterized by a stable proliferation arrest, and (iii) by a set of specific features. Timely regulated senescence is thought to be beneficial, whereas chronic senescence such as during normal or premature aging is deleterious as it favors most, if not all, age-related diseases. In this study, using in-house or publicly available microarray analyses of transcriptomes of senescent cells, as well as analyses of the level of expression of several DNA repair genes by RT-qPCR and immunoblot, we show that repression of DNA repair gene expression is associated with cellular senescence. This repression is mediated by the RB/E2F pathway and it may play a causal role in senescence induction, as single DNA repair gene repression by siRNA induced features of premature senescence. Importantly, activating RB independently of direct DNA damage also results in repression of DNA repair genes and in the subsequent induction of DNA damage and senescence. The dogma is that DNA damage observed during cellular senescence is directly provoked by DNA lesions following genotoxic attack (UV, IR, and ROS) or by induction of replicative stress upon oncogenic activation. Our in vitro results support a largely unsuspected contribution of the loss of DNA repair gene expression in the induction and the accumulation of the DNA damage observed in most, if not all, kinds of cellular senescence, and thus in the induction of cellular senescence. Further demonstration using in vivo models will help to generalize our findings.


Asunto(s)
Senescencia Celular , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Células Epiteliales/enzimología , Glándulas Mamarias Humanas/enzimología , Transcripción Genética , Células Cultivadas , Enzimas Reparadoras del ADN/genética , Regulación hacia Abajo , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Humanos , Glándulas Mamarias Humanas/citología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Nat Cell Biol ; 20(3): 272-284, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29459781

RESUMEN

For cancer cells to survive during extracellular matrix (ECM) detachment, they must inhibit anoikis and rectify metabolic deficiencies that cause non-apoptotic cell death. Previous studies in ECM-detached cells have linked non-apoptotic cell death to reactive oxygen species (ROS) generation, although the mechanistic underpinnings of this link remain poorly defined. Here, we uncover a role for receptor-interacting protein kinase 1 (RIPK1) in the modulation of ROS and cell viability during ECM detachment. We find that RIPK1 activation during ECM detachment results in mitophagy induction through a mechanism dependent on the mitochondrial phosphatase PGAM5. As a consequence of mitophagy, ECM-detached cells experience diminished NADPH production in the mitochondria, and the subsequent elevation in ROS levels leads to non-apoptotic death. Furthermore, we find that antagonizing RIPK1/PGAM5 enhances tumour formation in vivo. Thus, RIPK1-mediated induction of mitophagy may be an efficacious target for therapeutics aimed at eliminating ECM-detached cancer cells.


Asunto(s)
Células Epiteliales/enzimología , Matriz Extracelular/metabolismo , Glándulas Mamarias Humanas/enzimología , Mitocondrias/enzimología , Mitofagia , Neoplasias/enzimología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Adhesión Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Epiteliales/patología , Matriz Extracelular/patología , Femenino , Células HCT116 , Células HeLa , Humanos , Glándulas Mamarias Humanas/patología , Ratones Desnudos , Mitocondrias/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , NADP/metabolismo , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal , Carga Tumoral
12.
J Cell Physiol ; 233(3): 2464-2475, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28771726

RESUMEN

Annexin A2 (AnxA2) has been shown to play multiple roles in growth, development, and metabolism, but the functions of AnxA2 and the signaling pathways associated with AnxA2 are still not fully understood. In this study, we aim to reveal whether and how AnxA2 could be involved in milk synthesis and proliferation of bovine mammary epithelial cells (BMECs). Using gene function study approaches, we found that AnxA2 positively regulates PIP3 level, phosphorylation of mTOR, and protein levels of SREBP-1c and Cyclin D1 leading to milk synthesis and cell proliferation. We further observed that both AnxA2-36 kD phosphorylated form and AnxA2-33 kD protein could be induced from AnxA2-36 kD protein in BMECs under methionine, leucine, estrogen or prolactin stimulation. These above results strongly demonstrate that AnxA2 functions as a critical regulator for amino acid or hormone-induced milk synthesis and cell proliferation via the PI3K-mTOR-SREBP-1c/Cyclin D1 signaling pathway.


Asunto(s)
Anexina A2/metabolismo , Proliferación Celular , Células Epiteliales/enzimología , Glándulas Mamarias Humanas/enzimología , Proteínas de la Leche/biosíntesis , Serina-Treonina Quinasas TOR/metabolismo , Animales , Anexina A2/genética , Bovinos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclina D1/metabolismo , Células Epiteliales/efectos de los fármacos , Estrógenos/farmacología , Femenino , Humanos , Leucina/farmacología , Glándulas Mamarias Humanas/efectos de los fármacos , Metionina/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación , Progesterona/farmacología , Interferencia de ARN , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transfección
13.
Molecules ; 22(6)2017 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-28604588

RESUMEN

Docosahexaenoic acid (DHA), an ω-3 fatty acid abundant in fish oils, has diverse health beneficial effects, such as anti-oxidative, anti-inflammatory, neuroprotective, and chemopreventive activities. In this study, we found that DHA induced expression of two representative antioxidant/cytoprotective enzymes, heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase (NQO1), in human mammary epithealial (MCF-10A) cells. DHA-induced upregulation of these enzymes was accompanied by enhanced translocation of the redox-sensitive transcription factor Nrf2 into the nucleus and its binding to antioxidant response element. Nrf2 gene silencing by siRNA abolished the DHA-induced expression of HO-1 and NQO1 proteins. When MCF-10A cells were transfected with mutant constructs in which the cysteine 151 or 288 residue of Keap1 was replaced by serine, DHA-induced expression of HO-1 and NQO1 was markedly reduced. Moreover, DHA activated protein kinase C (PKC)δ and induced Nrf2 phosphorylation. DHA-induced phosphorylation of Nrf2 was abrogated by the pharmacological PKCδ inhibitor rottlerin or siRNA knockdown of its gene expression. The antioxidants N-acetyl-l-cysteine and Trolox attenuated DHA-induced activation of PKCδ, phosphorylation of Nrf2, and and its target protein expression. In conclusion, DHA activates Nrf2, possibly through modification of critical Keap1 cysteine 288 residue and PKCδ-mediated phosphorylation of Nrf2, leading to upregulation of HO-1 and NQO1 expression.


Asunto(s)
Ácidos Docosahexaenoicos/administración & dosificación , Hemo-Oxigenasa 1/genética , NAD(P)H Deshidrogenasa (Quinona)/genética , Factor 2 Relacionado con NF-E2/genética , Acetilcisteína/administración & dosificación , Antioxidantes/administración & dosificación , Cromanos/administración & dosificación , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/enzimología , Fosforilación/efectos de los fármacos , Proteína Quinasa C-delta/genética , ARN Interferente Pequeño/genética
14.
Cell Death Dis ; 8(5): e2769, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28492548

RESUMEN

Protein tyrosine phosphatase 1B (PTP1B) is a well-known inhibitor of insulin signaling pathways and inhibitors against PTP1B are being developed as promising drug candidates for treatment of obesity. PTP1B has also been linked to breast cancer both as a tumor suppressor and as an oncogene. Furthermore, PTP1B has been shown to be a regulator of cell adhesion and migration in normal and cancer cells. In this study, we analyzed the PTP1B expression in normal breast tissue, primary breast cells and the breast epithelial cell line D492. In normal breast tissue and primary breast cells, PTP1B is widely expressed in both epithelial and stromal cells, with highest expression in myoepithelial cells and fibroblasts. PTP1B is widely expressed in branching structures generated by D492 when cultured in 3D reconstituted basement membrane (3D rBM). Inhibition of PTP1B in D492 and another mammary epithelial cell line HMLE resulted in reduced cell proliferation and induction of anoikis. These changes were seen when cells were cultured both in monolayer and in 3D rBM. PTP1B inhibition affected cell attachment, expression of cell adhesion proteins and actin polymerization. Moreover, epithelial to mesenchymal transition (EMT) sensitized cells to PTP1B inhibition. A mesenchymal sublines of D492 and HMLE (D492M and HMLEmes) were more sensitive to PTP1B inhibition than D492 and HMLE. Reversion of D492M to an epithelial state using miR-200c-141 restored resistance to detachment induced by PTP1B inhibition. In conclusion, we have shown that PTP1B is widely expressed in the human breast gland with highest expression in myoepithelial cells and fibroblasts. Inhibition of PTP1B in D492 and HMLE affects cell-cell adhesion and induces anoikis-like effects. Finally, cells with an EMT phenotype are more sensitive to PTP1B inhibitors making PTP1B a potential candidate for further studies as a target for drug development in cancer involving the EMT phenotype.


Asunto(s)
Anoicis , Comunicación Celular , Células Epiteliales/enzimología , Regulación Enzimológica de la Expresión Génica , Glándulas Mamarias Humanas/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/biosíntesis , Adhesión Celular , Línea Celular , Femenino , Humanos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética
16.
Oncotarget ; 7(49): 80579-80585, 2016 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-27811358

RESUMEN

Lipid metabolism is crucially involved with the promotion of malignant progression and metastasis in various cancers. Growing evidence suggests that many types of cancers express high levels of sphingosine kinase 1 (Sphk1), which is known to mediate cell proliferation We hypothesized that Sphk1/sphingosine-1-phosphate (S1P) signaling contributes to tumor progression. In MCF10A and MCF10A-Sphk1 breast epithelial cells, we used TNF-α to activate the Sphk1/S1P pathway and the measured expression levels of NF-κBp65 and cyclin D1 mRNA and protein in the presence and absence of an NF-κB-p65 inhibitor. Chromatin immunoprecipitation assays were performed to determine whether NF-κB-p65 binds to the cyclin D1 promoter. We found that overexpression of Sphk1 induced NF-κB-p65 activation, increased expression of cyclin D1, shortened the cell division cycle, and thus promoted proliferation of breast epithelial cells. These findings provide insight into the mechanism by which an Sphk1/NF-κB-p65/cyclin D1 signaling pathway mediates cell proliferation.


Asunto(s)
Proliferación Celular , Ciclina D1/metabolismo , Células Epiteliales/enzimología , Glándulas Mamarias Humanas/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Factor de Transcripción ReIA/metabolismo , Sitios de Unión , Ciclo Celular , Línea Celular , Ciclina D1/genética , Células Epiteliales/patología , Femenino , Humanos , Lisofosfolípidos/metabolismo , Glándulas Mamarias Humanas/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Regiones Promotoras Genéticas , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Factores de Tiempo , Factor de Transcripción ReIA/genética , Transcripción Genética
17.
Oncotarget ; 7(50): 82273-82288, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27756891

RESUMEN

c-Myc is a potent driver of many human cancers. Since strategies for directly targeting c-Myc protein have had limited success, upstream regulators and downstream effectors of c-Myc are being investigated as alternatives for therapeutic intervention. c-Myc regulates transcription and formation of the mRNA cap, which is important for transcript maturation and translation. However, the direct mechanism by which c-Myc upregulates mRNA capping is unclear. mRNA cap formation initiates with the linkage of inverted guanosine via a triphosphate bridge to the first transcribed nucleotide, catalysed by mRNA capping enzyme (CE/RNGTT). Here we report that c-Myc increases the recruitment of catalytically active CE to RNA polymerase II and to its target genes. c-Myc-induced target gene expression, cell proliferation and cell transformation is highly dependent on CE, but only when c-Myc is deregulated. Cells retaining normal control of c-Myc expression are insensitive to repression of CE. c-Myc expression is also dependent on CE. Therefore, inhibiting CE provides an attractive route for selective therapeutic targeting of cancer cells which have acquired deregulated c-Myc.


Asunto(s)
Glándulas Mamarias Humanas/enzimología , Nucleotidiltransferasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Neoplasias del Cuello Uterino/enzimología , Sitios de Unión , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Nucleotidiltransferasas/genética , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Caperuzas de ARN/genética , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , Transfección , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
18.
Oncotarget ; 7(50): 81981-81994, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27626181

RESUMEN

Long non-coding RNAs (lncRNAs) have been implicated in normal cellular homeostasis as well as pathophysiological conditions, including cancer. Here we performed global gene expression profiling of mammary epithelial cells transformed by oncogenic v-Src, and identified a large subset of uncharacterized lncRNAs potentially involved in breast cancer development. Specifically, our analysis revealed a novel lncRNA, LINC00520 that is upregulated upon ectopic expression of oncogenic v-Src, in a manner that is dependent on the transcription factor STAT3. Similarly, LINC00520 is also increased in mammary epithelial cells transformed by oncogenic PI3K and its expression is decreased upon knockdown of mutant PIK3CA. Additional expression profiling highlight that LINC00520 is elevated in a subset of human breast carcinomas, with preferential enrichment in the basal-like molecular subtype. ShRNA-mediated depletion of LINC00520 results in decreased cell migration and loss of invasive structures in 3D. RNA sequencing analysis uncovers several genes that are differentially expressed upon ectopic expression of LINC00520, a significant subset of which are also induced in v-Src-transformed MCF10A cells. Together, these findings characterize LINC00520 as a lncRNA that is regulated by oncogenic Src, PIK3CA and STAT3, and which may contribute to the molecular etiology of breast cancer.


Asunto(s)
Neoplasias de la Mama/enzimología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Fosfatidilinositol 3-Quinasa Clase I/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Glándulas Mamarias Humanas/enzimología , Glándulas Mamarias Humanas/patología , Mutación , Invasividad Neoplásica , Proteína Oncogénica pp60(v-src)/genética , Proteína Oncogénica pp60(v-src)/metabolismo , Interferencia de ARN , ARN Largo no Codificante/genética , Factor de Transcripción STAT3/genética , Transducción de Señal , Factores de Tiempo , Transfección , Regulación hacia Arriba
19.
Oncotarget ; 7(39): 64109-64123, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27579892

RESUMEN

Several members of the Poly(ADP-ribose) polymerase (PARP) family are essential regulators of genome integrity, actively prospected as drug targets for cancer therapy. Among them, PARP3 is well characterized for its functions in double-strand break repair and mitotis. Here we report that PARP3 also plays an integral role in TGFß and reactive oxygen species (ROS) dependent epithelial-to-mesenchymal transition (EMT) and stem-like cell properties in human mammary epithelial and breast cancer cells. PARP3 expression is higher in breast cancer cells of the mesenchymal phenotype and correlates with the expression of the mesenchymal marker Vimentin while being in inverse correlation with the epithelial marker E-cadherin. Furthermore, PARP3 expression is significantly upregulated during TGFß-induced EMT in various human epithelial cells. In line with this observation, PARP3 depletion alters TGFß-dependent EMT of mammary epithelial cells by preventing the induction of the Snail-E-cadherin axis, the dissolution of cell junctions, the acquisition of cell motility and chemoresistance. PARP3 responds to TGFß-induced ROS to promote a TG2-Snail-E-cadherin axis during EMT. Considering the link between EMT and cancer stem cells, we show that PARP3 promotes stem-like cell properties in mammary epithelial and breast cancer cells by inducing the expression of the stem cell markers SOX2 and OCT4, by increasing the proportion of tumor initiating CD44high/CD24low population and the formation of tumor spheroid bodies, and by promoting stem cell self-renewal. These findings point to a novel role of PARP3 in the control of TGFß-induced EMT and acquisition of stem-like cell features and further motivate efforts to identify PARP3 specific inhibitors.


Asunto(s)
Neoplasias de la Mama/enzimología , Cadherinas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Transición Epitelial-Mesenquimal , Proteínas de Unión al GTP/metabolismo , Glándulas Mamarias Humanas/enzimología , Células Madre Neoplásicas/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Transglutaminasas/metabolismo , Células A549 , Antígenos CD , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Antígeno CD24/metabolismo , Cadherinas/genética , Proteínas de Ciclo Celular/genética , Movimiento Celular , Autorrenovación de las Células , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Etopósido/farmacología , Femenino , Proteínas de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Receptores de Hialuranos/metabolismo , Glándulas Mamarias Humanas/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fenotipo , Poli(ADP-Ribosa) Polimerasas/genética , Proteína Glutamina Gamma Glutamiltransferasa 2 , Interferencia de ARN , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Esferoides Celulares , Factores de Tiempo , Inhibidores de Topoisomerasa II/farmacología , Transfección , Transglutaminasas/genética
20.
Sci Rep ; 6: 25505, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27148852

RESUMEN

Regulation of Ca(2+) transport is vital in physiological processes, including lactation, proliferation and apoptosis. The plasmalemmal Ca(2+) pump isoform 2 (PMCA2) a calcium ion efflux pump, was the first protein identified to be crucial in the transport of Ca(2+) ions into milk during lactation in mice. In these studies we show that PMCA2 is also expressed in human epithelia undergoing lactational remodeling and also report strong PMCA2 staining on apical membranes of luminal epithelia in approximately 9% of human breast cancers we assessed. Membrane protein expression was not significantly associated with grade or hormone receptor status. However, PMCA2 mRNA levels were enriched in Basal breast cancers where it was positively correlated with survival. Silencing of PMCA2 reduced MDA-MB-231 breast cancer cell proliferation, whereas silencing of the related isoforms PMCA1 and PMCA4 had no effect. PMCA2 silencing also sensitized MDA-MB-231 cells to the cytotoxic agent doxorubicin. Targeting PMCA2 alone or in combination with cytotoxic therapy may be worthy of investigation as a therapeutic strategy in breast cancer. PMCA2 mRNA levels are also a potential tool in identifying poor responders to therapy in women with Basal breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Calcio/metabolismo , Carcinoma Basocelular/genética , Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Señalización del Calcio , Carcinoma Basocelular/tratamiento farmacológico , Carcinoma Basocelular/mortalidad , Carcinoma Basocelular/patología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Membrana Celular/patología , Proliferación Celular , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Doxorrubicina/farmacología , Células Epiteliales/enzimología , Células Epiteliales/patología , Femenino , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Lactancia/fisiología , Glándulas Mamarias Humanas/enzimología , Glándulas Mamarias Humanas/patología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/antagonistas & inhibidores , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...