Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1866(2): 290-299, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27750042

RESUMEN

Tumor microenvironment plays a crucial role in coordination with cancer cells in the establishment, growth and dissemination of the tumor. Among cells of the microenvironment, mesenchymal stem cells (MSCs) and their ability to evolve into cancer associated fibroblasts (CAFs) have recently generated a major interest in the field. Numerous studies have described the potential pro- or anti-tumorigenic action of MSCs. The goal of this review is to synthesize recent and emerging discoveries concerning the mechanisms by which MSCs can be attracted to tumor sites, how they can generate CAFs and by which way MSCs are able to modulate the growth, response to treatments, angiogenesis, invasion and metastasis of tumors. The understanding of the role of MSCs in tumor development has potential and clinical applications in terms of cancer management.


Asunto(s)
Comunicación Celular , Células Madre Mesenquimatosas/fisiología , Neoplasias/patología , Animales , Apoptosis , Fibroblastos Asociados al Cáncer/fisiología , Proliferación Celular , Humanos , Neoplasias/etiología , Neoplasias/metabolismo , Neovascularización Fisiológica , Microambiente Tumoral
2.
Trends Mol Med ; 30(1): 37-55, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37872025

RESUMEN

Recent findings have modified our understanding of the roles of chemokine receptor CXCR2 and its ligands in cancer, inflammation, and immunity. Studies in Cxcr2 tissue-specific knockout mice show that this receptor is involved in, among other things, cancer, central nervous system (CNS) function, metabolism, reproduction, COVID-19, and the response to circadian cycles. Moreover, CXCR2 involvement in neutrophil function has been revisited not only in physiology but also for its major contribution to cancers. The recent unfolding of the role of CXCR2 in numerous cancers has led to extensive evaluation of multiple CXCR2 antagonists in preclinical and clinical studies. In this review we discuss the potential of targeting CXCR2 for cancer treatment.


Asunto(s)
Neoplasias , Receptores de Interleucina-8B , Ratones , Animales , Humanos , Receptores de Interleucina-8B/genética , Inflamación/metabolismo , Neutrófilos , Neoplasias/genética , Neoplasias/metabolismo , Ratones Noqueados
3.
Nat Commun ; 14(1): 2058, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37045841

RESUMEN

WHIM Syndrome is a rare immunodeficiency caused by gain-of-function CXCR4 mutations. Here we report a decrease in bone mineral density in 25% of WHIM patients and bone defects leading to osteoporosis in a WHIM mouse model. Imbalanced bone tissue is observed in mutant mice combining reduced osteoprogenitor cells and increased osteoclast numbers. Mechanistically, impaired CXCR4 desensitization disrupts cell cycle progression and osteogenic commitment of skeletal stromal/stem cells, while increasing their pro-osteoclastogenic capacities. Impaired osteogenic differentiation is evidenced in primary bone marrow stromal cells from WHIM patients. In mice, chronic treatment with the CXCR4 antagonist AMD3100 normalizes in vitro osteogenic fate of mutant skeletal stromal/stem cells and reverses in vivo the loss of skeletal cells, demonstrating that proper CXCR4 desensitization is required for the osteogenic specification of skeletal stromal/stem cells. Our study provides mechanistic insights into how CXCR4 signaling regulates the osteogenic fate of skeletal cells and the balance between bone formation and resorption.


Asunto(s)
Síndromes de Inmunodeficiencia , Osteoporosis , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4 , Animales , Ratones , Síndromes de Inmunodeficiencia/genética , Mutación , Osteogénesis/genética , Osteoporosis/genética , Enfermedades de Inmunodeficiencia Primaria/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Humanos
4.
Front Pharmacol ; 13: 1095289, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36588678

RESUMEN

Activation of the aryl hydrocarbon receptor (AhR) through environmental exposure to chemicals including polycyclic aromatic hydrocarbons (PAHs) and polychlorinated dibenzo-p-dioxins (PCDDs) can lead to severe adverse health effects and increase the risk of breast cancer. This review considers several mechanisms which link the tumor promoting effects of environmental pollutants with the AhR signaling pathway, contributing to the development and progression of breast cancer. We explore AhR's function in shaping the tumor microenvironment, modifying immune tolerance, and regulating cancer stemness, driving breast cancer chemoresistance and metastasis. The complexity of AhR, with evidence for both oncogenic and tumor suppressor roles is discussed. We propose that AhR functions as a "molecular bridge", linking disproportionate toxin exposure and policies which underlie environmental injustice with tumor cell behaviors which drive poor patient outcomes.

5.
Front Immunol ; 13: 1005551, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36311783

RESUMEN

Neutrophils play a major role in the protection from infections but also in inflammation related to tumor microenvironment. However, cell-extrinsic and -intrinsic cues driving their function at steady state is still fragmentary. Using Cxcr2 knock-out mice, we have evaluated the function of the chemokine receptor Cxcr2 in neutrophil physiology. We show here that Cxcr2 deficiency decreases the percentage of mature neutrophils in the spleen, but not in the bone marrow (BM). There is also an increase of aged CD62Llo CXCR4hi neutrophils in the spleen of KO animals. Spleen Cxcr2-/- neutrophils display a reduced phagocytic ability, whereas BM neutrophils show an enhanced phagocytic ability compared to WT neutrophils. Spleen Cxcr2-/- neutrophils show reduced reactive oxygen species production, F-actin and α-tubulin levels. Moreover, spleen Cxcr2-/- neutrophils display an altered signaling with reduced phosphorylation of ERK1/2 and p38 MAPK, impaired PI3K-AKT, NF-κB, TGFß and IFNγ pathways. Altogether, these results suggest that Cxcr2 is essential for neutrophil physiology.


Asunto(s)
Neutrófilos , Fosfatidilinositol 3-Quinasas , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo
6.
Arch Biochem Biophys ; 512(1): 78-86, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21640702

RESUMEN

The aryl hydrocarbon receptor (AhR) has been best known for its role in mediating the toxicity of dioxin. Here we show that AhR overexpression is found among estrogen receptor (ER)α-negative human breast tumors and that its overexpression is positively correlated to that of the NF-κB subunit RelB and Interleukin (IL)-8. Increased DNA binding activity of the AhR and RelB is coupled to IL-8 overexpression in primary breast cancer tissue, which was also supported by in situ hybridization. Activation of AhR in vitro by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced IL-8 expression in MDA-MB 436 and MCF-7 cells in an AhR and RelB dependent manner. Consistently, downregulation of RelB or AhR by small interfering RNAs (siRNA) decreased the level of IL-8 but increased expression of ERα in vitro in MCF-7 cells. Our results strongly suggest that RelB and AhR have a critical role in the regulation of IL-8 and reveal a supportive role of RelB and AhR in the anti-apoptotic response in human breast cancer cells. AhR and RelB may present a novel therapeutic target for inflammatory driven breast carcinogenesis and tumor progression. Overexpression of pro-survival factors AhR and RelB may explain the process of the development of environmentally-induced type of breast cancers.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Interleucina-8/genética , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción ReIB/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Cultivadas , ADN/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Interleucina-8/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Unión Proteica , Receptores de Hidrocarburo de Aril/genética , Factor de Transcripción ReIB/genética , Regulación hacia Arriba
7.
Med Sci (Paris) ; 27(3): 285-8, 2011 Mar.
Artículo en Francés | MEDLINE | ID: mdl-21447301

RESUMEN

Mesenchymal stem cells (MSC) have attracted recent attention for their cell therapy potential, based in particular on their immunosuppressive properties, which have served as the basis for the treatment of autoimmune diseases. Interestingly, MSC have been used in cell therapy strategies to deliver therapeutical genes. Cell therapy approaches taking advantages of MSC have been proposed, as MSC display a potential tropsim for tumors. However, all these strategies raise a series of questions about the safety of MSC, as MSC could enhance tumor growth and metastasis. This review summarizes recent findngs about MSC in carcinogenesis.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/efectos adversos , Neoplasias/cirugía , Humanos , Células Madre Mesenquimatosas/fisiología , Neoplasias/patología , Tropismo
8.
Cancers (Basel) ; 13(10)2021 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-34066060

RESUMEN

Chemokines and their receptors are key players in breast cancer progression and outcome. Previous studies have shown that the chemokine receptor CXCR2 was expressed at higher levels by cells of the tumor microenvironment in triple-negative breast cancers (TNBCs). The aim of this study was to focus our attention on a retrospective cohort of 290 TNBC cases and analyze the involvement of CXCR2, CD11b (a marker of granulocytes) and CD66b (a marker of neutrophils) and their link with immune infiltration and immune checkpoint markers. We report that high densities of CXCR2-, CD11b- and CD66b-positive cells were associated with high-grade tumors. Moreover, molecular apocrine TNBCs, defined here as tumors that express both AR and FOXA1 biomarkers, exhibited low levels of CXCR2 and CD11b. High CXCR2 and CD11b levels were correlated with elevated density of tumor-infiltrating lymphocytes (TILs), CD8+ cytotoxic lymphocytes, expression of PD-L1 by tumor and stromal cells and of PD-1 by stromal cells. On the other hand, CD66b levels were associated only with CD8+, stromal PD-L1 and PD-1 expression. In univariate analysis, low levels of CXCR2 were correlated with poor OS and RFS. In multivariate analysis, low levels of CXCR2 were associated with poor OS. Finally, in TNBC treated with adjuvant chemotherapy, CXCR2 density was associated with longer RFS. Overall, our data highlight the potential beneficial association of high levels of CXCR2 with a subgroup of TNBC patients characterized by a better prognosis.

9.
Front Immunol ; 12: 625346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33763068

RESUMEN

Activation of the aryl hydrocarbon receptor (AhR) through environmental exposure to known human carcinogens including dioxins can lead to the promotion of breast cancer. While the repressor protein of the AhR (AhRR) blocks the canonical AhR pathway, the function of AhRR in the development of breast cancer is not well-known. In the current study we examined the impact of suppressing AhR activity using its dedicated repressor protein AhRR. AhRR is a putative tumor suppressor and is silenced in several cancer types, including breast, where its loss correlates with shorter patient survival. Using the AhRR transgenic mouse, we demonstrate that AhRR overexpression opposes AhR-driven and inflammation-induced growth of mammary tumors in two different murine models of breast cancer. These include a syngeneic model using E0771 mammary tumor cells as well as the Polyoma Middle T antigen (PyMT) transgenic model. Further AhRR overexpression or knockout of AhR in human breast cancer cells enhanced apoptosis induced by chemotherapeutics and inhibited the growth of mouse mammary tumor cells. This study provides the first in vivo evidence that AhRR suppresses mammary tumor development and suggests that strategies which lead to its functional restoration and expression may have therapeutic benefit.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/metabolismo , Transformación Celular Neoplásica/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Animales , Animales Modificados Genéticamente , Antígenos Transformadores de Poliomavirus/genética , Antineoplásicos/farmacología , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Etopósido/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ratones Endogámicos C57BL , Receptores de Hidrocarburo de Aril/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Carga Tumoral , Células Tumorales Cultivadas
10.
Cancers (Basel) ; 13(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070438

RESUMEN

Chemokines present in the tumor microenvironment are essential for the control of tumor progression. We show here that several ligands of the chemokine receptor Cxcr2 were up-regulated in the PyMT (polyoma middle T oncogene) model of breast cancer. Interestingly, the knock-down of Cxcr2 in PyMT animals led to an increased growth of the primary tumor and lung metastasis. The analysis of tumor content of PyMT-Cxcr2-/- animals highlighted an increased infiltration of tumor associated neutrophils (TANs), mirrored by a decreased recruitment of tumor associated macrophages (TAMs) compared to PyMT animals. Analysis of PyMT-Cxcr2-/- TANs revealed that they lost their killing ability compared to PyMT-Cxcr2+/+ TANs. The transcriptomic analysis of PyMT-Cxcr2-/- TANs showed that they had a more pronounced pro-tumor TAN2 profile compared to PyMT TANs. In particular, PyMT-Cxcr2-/- TANs displayed an up-regulation of the pathways involved in reactive oxygen species (ROS) production and angiogenesis and factors favoring metastasis, but reduced apoptosis. In summary, our data reveal that a lack of Cxcr2 provides TANs with pro-tumor effects.

11.
Front Immunol ; 12: 766275, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858421

RESUMEN

Knowledge about the hematopoietic niche has evolved considerably in recent years, in particular through in vitro analyzes, mouse models and the use of xenografts. Its complexity in the human bone marrow, in particular in a context of hematological malignancy, is more difficult to decipher by these strategies and could benefit from the knowledge acquired on the niches of solid tumors. Indeed, some common features can be suspected, since the bone marrow is a frequent site of solid tumor metastases. Recent research on solid tumors has provided very interesting information on the interactions between tumoral cells and their microenvironment, composed notably of mesenchymal, endothelial and immune cells. This review thus focuses on recent discoveries on tumor niches that could help in understanding hematopoietic niches, with special attention to 4 particular points: i) the heterogeneity of carcinoma/cancer-associated fibroblasts (CAFs) and mesenchymal stem/stromal cells (MSCs), ii) niche cytokines and chemokines, iii) the energy/oxidative metabolism and communication, especially mitochondrial transfer, and iv) the vascular niche through angiogenesis and endothelial plasticity. This review highlights actors and/or pathways of the microenvironment broadly involved in cancer processes. This opens avenues for innovative therapeutic opportunities targeting not only cancer stem cells but also their regulatory tumor niche(s), in order to improve current antitumor therapies.


Asunto(s)
Microambiente Tumoral , Animales , Fibroblastos Asociados al Cáncer , Comunicación Celular , Citocinas , Endotelio Vascular , Humanos , Células Madre Mesenquimatosas , Neoplasias , Neovascularización Patológica
12.
Cancers (Basel) ; 12(8)2020 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-32727083

RESUMEN

The tumor microenvironment appears essential in cancer progression and chemokines are mediators of the communication between cancer cells and stromal cells. We have previously shown that the ligands of the chemokine receptor CXCR2 were expressed at higher levels in triple-negative breast cancers (TNBC). Our hypothesis was that CXCR2 expression could also be altered in breast cancer. Here, we have analyzed the potential role of CXCR2 in breast cancer in a retrospective cohort of 105 breast cancer patients. Expression of CXCR2, CD11b (a marker of granulocytes) and CD66b (a marker of neutrophils) was analyzed by immunohistochemistry on tumor samples. We demonstrated that CXCR2 stained mainly stromal cells and in particular neutrophils. CXCR2, CD11b and CD66b expression were correlated with high grade breast cancers. Moreover, TNBC displayed a higher expression of CXCR2, CD11b and CD66b than hormone receptor positive or Her2 positive tumors. High levels of CXCR2 and CD11b, but not CD66b, were associated with a higher infiltration of T lymphocytes and B lymphocytes. We also observed a correlation between CXCR2 and AP-1 activity. In univariate analyses, CXCR2, but not CD11b or CD66b, was associated with a lower risk of relapse; CXCR2 remained significant in multivariate analysis. Our data suggest that CXCR2 is a stromal marker of TNBC. However, higher levels of CXCR2 predicted a lower risk of relapse.

13.
Life Sci Alliance ; 3(3)2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32041848

RESUMEN

Microbiota and chronic infections can affect not only immune status, but also the overall physiology of animals. Here, we report that chronic infections dramatically modify the phenotype of Cxcr2 KO mice, impairing in particular, their reproduction ability. We show that exposure of Cxcr2 KO females to multiple types of chronic infections prevents their ability to cycle, reduces the development of the mammary gland and alters the morphology of the uterus due to an impairment of ovary function. Mammary gland and ovary transplantation demonstrated that the hormonal contexture was playing a crucial role in this phenomenon. This was further evidenced by alterations to circulating levels of sex steroid and pituitary hormones. By analyzing at the molecular level the mechanisms of pituitary dysfunction, we showed that in the absence of Cxcr2, bystander infections affect leukocyte migration, adhesion, and function, as well as ion transport, synaptic function behavior, and reproduction pathways. Taken together, these data reveal that a chemokine receptor plays a direct role in pituitary function and reproduction in the context of chronic infections.


Asunto(s)
Hipófisis/fisiología , Receptores de Interleucina-8B/metabolismo , Animales , Femenino , Hormonas Esteroides Gonadales/metabolismo , Infecciones/microbiología , Infecciones/fisiopatología , Masculino , Ratones , Ratones Endogámicos BALB C , Ovario/metabolismo , Enfermedades de la Hipófisis/metabolismo , Hipófisis/metabolismo , Receptores de Interleucina-8B/genética , Reproducción , Útero/metabolismo
14.
Int J Cancer ; 124(10): 2270-80, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19173296

RESUMEN

Elevated deoxycholic acid (DCA), mutations in the adenomatous polyposis coli (APC) gene and chronic inflammation are associated with increased risk of colorectal cancer. APC status was manipulated to determine whether DCA mediates inflammatory molecules in normal or initiated colonic mucosa. DCA increased steady state mRNA and protein levels of CXCL8 in cells which do not express wild-type APC. Steady-state CXCL8 mRNA and protein were suppressed when cells with conditional expression of wild-type APC were exposed to DCA. Immunostaining did not detect CXCL8 in normal human colonic mucosa. CXCL8 was expressed in adenomatous polyps and adenocarcinomas. CXCL8 expression correlated with nuclear beta-catenin localization in epithelial cells of adenomas, but was associated with endothelial cells and neutrophils in the adenocarcinomas. DCA-mediated CXCL8 promoter-reporter activity was elevated in a mutant APC background. Wild-type APC suppressed this effect. Mutation of activator protein-1 (AP-1) or nuclear factor kappa B (NF-kappaB) sites suppressed the activation of the CXCL8 promoter-reporter by DCA. Chromatin immunoprecipitation revealed that AP-1 and NF-kappaB binding to the 5'-promoter of CXCL8 was induced by DCA. The beta-catenin transcription factor was bound to the 5'-promoter of CXCL8 in the absence or presence of DCA. Phenotypic assays determined that DCA-mediated invasion was blocked by antibody-directed against CXCL8 or wild-type APC. CXCL8 exposure led to matrix metalloproteinase-2 production and increased invasion on laminin-coated filters. These data suggest that DCA-mediated CXCL8 occurs in initiated colonic epithelium and neutralizing CXCL8 could reduce the invasive potential of tumors.


Asunto(s)
Neoplasias Colorrectales/genética , Ácido Desoxicólico/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes APC , Interleucina-8/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cartilla de ADN , Humanos , Inmunohistoquímica , Interleucina-8/genética , ARN Mensajero/genética , Transcripción Genética/efectos de los fármacos
15.
Stem Cells ; 26(6): 1387-94, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18388305

RESUMEN

This review focuses on the interaction between multipotent stromal cells (MSCs) and carcinoma and the possible use of MSCs in cell-based anticancer therapies. MSCs are present in multiple tissues and are defined as cells displaying the ability to differentiate in multiple lineages, including chondrocytes, osteoblasts, and adipocytes. Recent evidence also suggests that they could play a role in the progression of carcinogenesis and that MSCs could migrate toward primary tumors and metastatic sites. It is possible that MSCs could also be involved in the early stages of carcinogenesis through spontaneous transformation. In addition, it is thought that MSCs can modulate tumor growth and metastasis, although this issue remains controversial and not well understood. The immunosuppressive properties and proangiogenic properties of MSCs account, at least in part, for their effects on cancer development. On the other hand, cancer cells also have the ability to enhance MSC migration. This complex dialog between MSCs and cancer cells is certainly critical for the outcome of tumor development. Interestingly, several studies have shown that MSCs engineered to express antitumor factors could be an innovative choice as a cell-mediated gene therapy to counteract tumor growth. More evidence will be needed to understand how MSCs positively or negatively modulate carcinogenesis and to evaluate the safety of MSC use in cell-mediated gene strategies. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Neoplasias/epidemiología , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre/efectos adversos , Células del Estroma/trasplante , Adulto , Animales , Transformación Celular Neoplásica , Humanos , Terapia de Inmunosupresión , Modelos Animales , Neovascularización Patológica , Medición de Riesgo
16.
Mol Endocrinol ; 22(2): 317-30, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17962381

RESUMEN

The ubiquitin-proteasome pathway has been recognized as an important regulator in the hormonal response by estrogen receptor (ER) alpha, but its impact on ERbeta function is poorly characterized. In the current study, we investigated the role of the ubiquitin-proteasome pathway in regulating ERbeta activity and identified regulatory sites within the activation function (AF)-1 domain that modulate ERbeta ubiquitination and nuclear dynamics in a hormone-independent manner. Although both ERalpha and ERbeta were dependent on proteasome function for their maximal response to estrogen, they were regulated differently by proteasome inhibition in the absence of hormone, an effect shown to be dependent on their respective AF-1 domain. Given the role of AF-1 phosphorylation to regulate ER activity, we found that sequential substitutions of specific serine residues contained in MAPK consensus sites conferred transcriptional activation of ERbeta in a proteasome-dependent manner through reduced ubiquitination and enhanced accumulation of mutant receptors. Specifically, serines 94 and 106 within ERbeta AF-1 domain were found to modulate subnuclear mobility of the receptor to transit between inactive clusters and a more mobile state in a proteasome-dependent manner. In addition, cellular levels of ERbeta were regulated through these sites by facilitating the recruitment of the ubiquitin ligase E6-associated protein in a phosphorylation-dependent manner. These findings suggest a role for ERbeta AF-1 in contributing to the activation-degradation cycling of the receptor through a functional clustering of phosphorylated serine residues that cooperate in generating signals to the ubiquitin-proteasome pathway.


Asunto(s)
Núcleo Celular/metabolismo , Receptor beta de Estrógeno/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Sitios de Unión/genética , Línea Celular , Secuencia de Consenso , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Immunoblotting , Inmunoprecipitación , Leupeptinas/farmacología , Ratones , Modelos Biológicos , Mutación , Fosforilación , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma , Serina/genética , Serina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
17.
Trends Mol Med ; 30(1): 3-5, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38211559
18.
Mol Pharmacol ; 74(5): 1359-66, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18669446

RESUMEN

We have reported recently that the chemokine interleukin 8 (IL-8)/CXCL8 was overexpressed in invasive estrogen receptor (ERalpha)-negative breast cancer cells compared with ERalpha-positive breast cancer cells. We now demonstrate that histone deacetylases (HDACs) play an essential role in the regulation of IL-8 gene expression in ERalpha-positive MCF-7 breast cancer cells. Treatment of MCF-7 cells with the HDAC inhibitor trichostatin A (TSA) led to a strong up-regulation of IL-8 protein and RNA levels in MCF-7 cells. The up-regulation of IL-8 in MCF-7 cells was time- and concentration-dependent. Moreover, run-on and transfection experiments demonstrated that IL-8 induction by HDAC inhibitors was transcriptional and involved mainly the nuclear factor-kappaB (NF-kappaB) site of the IL-8 promoter. These observations are corroborated by an up-regulation of NF-kappaB activity in MCF-7 cells in the presence of TSA. In addition, blocking NF-kappaB pathway by adenoviral delivery of a dominant-negative IkappaBorIkappaB kinase complex 2 (IKK2) mutant abolished IL-8 gene induction by histone deacetylase inhibitors. HDAC inhibitors triggered IKK phosphorylation and up-regulated p65 nuclear translocation, although they decreased the protein levels of IkappaBalpha, which accounts for NF-kappaB activation. TSA increased binding of acetylated histone 3 to the IL-8 gene promoter. In summary, our results demonstrate that NF-kappaB pathway repression by HDAC is responsible for the low expression of IL-8 in ERalpha-positive breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Histona Desacetilasas/metabolismo , Interleucina-8/metabolismo , FN-kappa B/metabolismo , Adenoviridae/genética , Secuencia de Bases , Neoplasias de la Mama/patología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-8/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , Recombinación Genética , Transducción de Señal
19.
Mol Endocrinol ; 21(12): 2941-55, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17823304

RESUMEN

The nuclear factor-kappaB (NF-kappaB) transcription factor family has a crucial role in rapid responses to stress and pathogens. We show that the NF-kappaB subunit RelB is functionally associated with the aryl hydrocarbon receptor (AhR) and mediates transcription of chemokines such as IL-8 via activation of AhR and protein kinase A. RelB physically interacts with AhR and binds to an unrecognized RelB/AhR responsive element of the IL-8 promoter linking two signaling pathways to activate gene transcription. We found a time-dependent recruitment of AhR to the RelB/AhR responsive element site of IL-8 mediated by the AhR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin) and via activation of protein kinase A. Furthermore, NF-kappaB-binding sites that are preferentially recognized by RelB/p52 are a target for RelB/AhR complexes without addition of any stimuli, implicating the endogenous function of the AhR. RelB/AhR complexes are also found to bind on xenobiotic responsive element, and RelB drastically increases the 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced xenobiotic responsive element reporter activity. The interaction of RelB with AhR signaling, and AhR with NF-kappaB RelB signaling pathways represent a new mechanism of cross talk between the two transcription factors.


Asunto(s)
Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción ReIB/metabolismo , Transcripción Genética/genética , Sitios de Unión , Línea Celular Tumoral , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Dibenzodioxinas Policloradas/farmacología , Regiones Promotoras Genéticas , Unión Proteica , Receptores de Hidrocarburo de Aril/genética , Elementos de Respuesta , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIB/genética
20.
Endocr Relat Cancer ; 14(4): 1039-52, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18045955

RESUMEN

Recent data suggest that chemokines could be essential players in breast carcinogenesis. We previously showed that the CXC chemokine CXCL8 (interleukin-8) was overexpressed in estrogen receptor alpha (ERalpha)-negative breast cell lines. Analysis of CXCL8 chromosomal location showed that several CXC chemokines (CXCL1, CXCL2, CXCL3, CXCL4, CXCL4V1, CXCL5, CXCL6, CXCL7, and CXCL8) were localized in the same narrow region (360 kb in size) of chromosome 4. We thus hypothesized that they could belong to the same cluster. Quantification of these chemokines in breast tumors showed that samples expressing high CXCL8 also produced elevated levels of CXCL1, CXCL3, and CXCL5, and displayed low content of ERalpha. CXCL1, CXCL2, CXCL3, CXCL5, and CXCL8 were co-regulated both in tumors and in breast cancer cell lines. CXCL5 and CXCL8 were mainly produced by epithelial cells, whereas CXCL1, CXCL2, and CXCL3 had a high expression in blood cells. The overexpression of these chemokines in tumor cells was not the result of gene amplification, but rather of an enhanced gene transcription. Our data suggest that high CXCL8 expression in tumors is mainly correlated to activating protein-1 (AP-1) pathway and to a minor extent to NF-kappaB pathway. Interestingly, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, and CXCL8 chemokines were present at higher levels in metastases when compared with grade I and III biopsies. High levels of CXCL8, CXCL1, and CXCL3 accounted for a shorter relapse-free survival of ERalpha-positive patients treated with tamoxifen. In summary, we present evidences that multiple CXC chemokines are co-expressed in CXCL8-positive breast tumors. In addition, these chemokines could account for the higher aggressiveness of these types of tumors.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Quimiocinas CXC/genética , Cromosomas Humanos Par 4 , Regulación Neoplásica de la Expresión Génica , Regulación hacia Arriba , Anciano , Neoplasias de la Mama/cirugía , Células Cultivadas , Mapeo Cromosómico , Supervivencia sin Enfermedad , Endotelio Vascular , Femenino , Humanos , Metástasis Linfática , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Venas Umbilicales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA