Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Theranostics ; 14(1): 249-264, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38164151

RESUMEN

Rationale: 17ß-estradiol (E2) can directly promote the growth of ERα-negative cancer cells through activation of endothelial ERα in the tumor microenvironment, thereby increasing a normalized tumor angiogenesis. ERα acts as a transcription factor through its nuclear transcriptional AF-1 and AF-2 transactivation functions, but membrane ERα plays also an important role in endothelium. The present study aims to decipher the respective roles of these two pathways in ERα-negative tumor growth. Moreover, we delineate the actions of tamoxifen, a Selective Estrogen Receptor Modulator (SERM) in ERα-negative tumors growth and angiogenesis, since we recently demonstrated that tamoxifen impacts vasculature functions through complex modulation of ERα activity. Methods: ERα-negative B16K1 cancer cells were grafted into immunocompetent mice mutated for ERα-subfunctions and tumor growths were analyzed in these different models in response to E2 and/or tamoxifen treatment. Furthermore, RNA sequencings were analyzed in endothelial cells in response to these different treatments and validated by RT-qPCR and western blot. Results: We demonstrate that both nuclear and membrane ERα actions are required for the pro-tumoral effects of E2, while tamoxifen totally abrogates the E2-induced in vivo tumor growth, through inhibition of angiogenesis but promotion of vessel normalization. RNA sequencing indicates that tamoxifen inhibits the E2-induced genes, but also initiates a specific transcriptional program that especially regulates angiogenic genes and differentially regulates glycolysis, oxidative phosphorylation and inflammatory responses in endothelial cells. Conclusion: These findings provide evidence that tamoxifen specifically inhibits angiogenesis through a reprogramming of endothelial gene expression via regulation of some transcription factors, that could open new promising strategies to manage cancer therapies affecting the tumor microenvironment of ERα-negative tumors.


Asunto(s)
Neoplasias , Tamoxifeno , Ratones , Animales , Tamoxifeno/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Células Endoteliales/metabolismo , Angiogénesis , Expresión Génica , Endotelio/metabolismo , Línea Celular Tumoral , Microambiente Tumoral/genética
2.
Arterioscler Thromb Vasc Biol ; 40(9): 2143-2158, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32640903

RESUMEN

OBJECTIVE: ERα (estrogen receptor alpha) exerts nuclear genomic actions and also rapid membrane-initiated steroid signaling. The mutation of the cysteine 451 into alanine in vivo has recently revealed the key role of this ERα palmitoylation site on some vasculoprotective actions of 17ß-estradiol (E2) and fertility. Here, we studied the in vivo role of the arginine 260 of ERα which has also been described to be involved in its E2-induced rapid signaling with PI-3K (phosphoinositide 3-kinase) as well as G protein in cultured cell lines. Approach and Results: We generated a mouse model harboring a point mutation of the murine counterpart of this arginine into alanine (R264A-ERα). In contrast to the C451A-ERα, the R264A-ERα females are fertile with standard hormonal serum levels and normal control of hypothalamus-pituitary ovarian axis. Although R264A-ERα protein abundance was normal, the well-described membrane ERα-dependent actions of estradiol, such as the rapid dilation of mesenteric arteries and the acceleration of endothelial repair of carotid, were abrogated in R264A-ERα mice. In striking contrast, E2-regulated gene expression was highly preserved in the uterus and the aorta, revealing intact nuclear/genomic actions in response to E2. Consistently, 2 recognized nuclear ERα-dependent actions of E2, namely atheroma prevention and flow-mediated arterial remodeling were totally preserved. CONCLUSIONS: These data underline the exquisite role of arginine 264 of ERα for endothelial membrane-initiated steroid signaling effects of E2 but not for nuclear/genomic actions. This provides the first model of fertile mouse with no overt endocrine abnormalities with specific loss-of-function of rapid ERα signaling in vascular functions.


Asunto(s)
Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Endotelio Vascular/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/agonistas , Terapia de Reemplazo de Estrógeno , Estrógenos/farmacología , Fertilidad/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Mutación Puntual , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/prevención & control , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/fisiopatología , Proliferación Celular/efectos de los fármacos , Endotelio Vascular/lesiones , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Activación Enzimática , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Ciclo Estral/efectos de los fármacos , Femenino , Masculino , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ovariectomía , Repitelización/efectos de los fármacos , Transducción de Señal , Factores de Tiempo , Útero/efectos de los fármacos , Útero/metabolismo , Remodelación Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos
4.
Development ; 147(5)2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32098763

RESUMEN

17ß-Estradiol induces the postnatal development of mammary gland and influences breast carcinogenesis by binding to the estrogen receptor ERα. ERα acts as a transcription factor but also elicits rapid signaling through a fraction of ERα expressed at the membrane. Here, we have used the C451A-ERα mouse model mutated for the palmitoylation site to understand how ERα membrane signaling affects mammary gland development. Although the overall structure of physiological mammary gland development is slightly affected, both epithelial fragments and basal cells isolated from C451A-ERα mammary glands failed to grow when engrafted into cleared wild-type fat pads, even in pregnant hosts. Similarly, basal cells purified from hormone-stimulated ovariectomized C451A-ERα mice did not produce normal outgrowths. Ex vivo, C451A-ERα basal cells displayed reduced matrix degradation capacities, suggesting altered migration properties. More importantly, C451A-ERα basal cells recovered in vivo repopulating ability when co-transplanted with wild-type luminal cells and specifically with ERα-positive luminal cells. Transcriptional profiling identified crucial paracrine luminal-to-basal signals. Altogether, our findings uncover an important role for membrane ERα expression in promoting intercellular communications that are essential for mammary gland development.


Asunto(s)
Epitelio/metabolismo , Receptor alfa de Estrógeno/biosíntesis , Glándulas Mamarias Animales/embriología , Comunicación Paracrina/fisiología , Animales , Células Epiteliales/metabolismo , Células Epiteliales/trasplante , Estradiol/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Lipoilación/fisiología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
5.
Sci Rep ; 9(1): 20169, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882815

RESUMEN

Metabolic diseases such as obesity, type II diabetes and hepatic steatosis are a public health concern in developed countries. The metabolic risk is gender-dependent. The constitutive androstane receptor (CAR), which is at the crossroads between energy metabolism and endocrinology, has recently emerged as a promising therapeutic agent for the treatment of obesity and type 2 diabetes. In this study we sought to determine its role in the dimorphic regulation of energy homeostasis. We tracked male and female WT and CAR deficient (CAR-/-) mice for over a year. During aging, CAR-/- male mice developed hypercortisism, obesity, glucose intolerance, insulin insensitivity, dyslipidemia and hepatic steatosis. Remarkably, the latter modifications were absent, or minor, in female CAR-/- mice. When ovariectomized, CAR-/- female mice developed identical patterns of metabolic disorders as observed in male mice. These results highlight the importance of steroid hormones in the regulation of energy metabolism by CAR. They unveil a sexually dimorphic role of CAR in the maintenance of endocrine and metabolic homeostasis underscoring the importance of considering sex in treatment of metabolic diseases.

6.
Arterioscler Thromb Vasc Biol ; 38(6): 1346-1357, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29650694

RESUMEN

OBJECTIVE: Estrogens exert beneficial effect on the blood vascular system. However, their role on the lymphatic system has been poorly investigated. We studied the protective effect of the 17ß estradiol-the most potent endogenous estrogen-in lymphedema-a lymphatic dysfunction, which results in a massive fluid and fat accumulation in the limb. APPROACH AND RESULTS: Screening of DNA motifs able to mobilize ERs (estrogen receptors) and quantitative real-time polymerase chain reaction analysis revealed that estradiol promotes transcriptional activation of lymphangiogenesis-related gene expression including VEGF (vascular endothelial growth factor)-D, VEGFR (VEGF receptor)-3, lyve-1, and HASs (hyaluronan synthases). Using an original model of secondary lymphedema, we observed a protective effect of estradiol on lymphedema by reducing dermal backflow-a representative feature of the pathology. Blocking ERα by tamoxifen-the selective estrogen modulator-led to a remodeling of the lymphatic network associated with a strong lymphatic leakage. Moreover, the protection of lymphedema by estradiol treatment was abrogated by the endothelial deletion of the receptor ERα in Tie2-Cre; ERαlox/lox mice, which exhibit dilated lymphatic vessels. This remodeling correlated with a decrease in lymphangiogenic gene expression. In vitro, blocking ERα by tamoxifen in lymphatic endothelial cells decreased cell-cell junctions, inhibited migration and sprouting, and resulted in an inhibition of Erk but not of Akt phosphorylation. CONCLUSIONS: Estradiol protection from developing lymphedema is mediated by an activation of its receptor ERα and is antagonized by tamoxifen. These findings reveal a new facet of the estrogen influence in the management of the lymphatic system and provide more evidence that secondary lymphedema is worsened by hormone therapy.


Asunto(s)
Linfedema del Cáncer de Mama/prevención & control , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/agonistas , Terapia de Reemplazo de Hormonas , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Linfedema del Cáncer de Mama/metabolismo , Linfedema del Cáncer de Mama/patología , Linfedema del Cáncer de Mama/fisiopatología , Modelos Animales de Enfermedad , Implantes de Medicamentos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Vasos Linfáticos/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía , Fosforilación , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Tamoxifeno/toxicidad
7.
Med Sci (Paris) ; 34(12): 1056-1062, 2018 Dec.
Artículo en Francés | MEDLINE | ID: mdl-30623764

RESUMEN

Women now live more than a third of their lives after the onset of menopause. The decline in endogenous estrogen production during this period is accompanied by functional disorders that affect quality of life. These symptoms may be relieved by menopausal hormone therapy (MHT) initially based on the administration of equine conjugated estrogens (mainly in the United States, oral route) or the natural estrogen, 17ß-estradiol (in Europe, transdermal route). Estrogen receptor α (ERα), but not ERß, mediates most of the physiological effects of estrogens. ERα belongs to the superfamily of nuclear receptors and regulates the transcription of genes via its activation functions AF1 and AF2. In addition to these classical genomic actions, estrogens can activate a subpopulation of ERα present at the cell membrane and thereby induce rapid signals. In this review, we will summarize the evolution of MHTs in last decades, as well as treatments that use various selective estrogen receptor modulators (SERMs). Next, we will describe recent advances in the understanding of the mechanisms of estrogen action, in particular the respective roles of nuclear and membrane ERα as well as the potential implications for future therapies.


Asunto(s)
Terapia de Reemplazo de Estrógeno/tendencias , Menopausia/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Animales , Calibración , Receptor alfa de Estrógeno/metabolismo , Terapia de Reemplazo de Estrógeno/métodos , Terapia de Reemplazo de Estrógeno/normas , Femenino , Humanos , Menopausia/fisiología , Calidad de Vida , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación
8.
Arterioscler Thromb Vasc Biol ; 37(5): 909-919, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28360090

RESUMEN

OBJECTIVE: Chronic nonhealing wounds are a substantial medical concern and are associated with morbidity and mortality; thus, new treatment strategies are required. The first step toward personalized/precision medicine in this field is probably in taking sex differences into account. Impaired wound healing is augmented by ischemia, and we previously demonstrated that 17ß-estradiol exerts a major preventive effect against ischemia-induced skin flap necrosis in female mice. However, the equivalent effects of testosterone in male mice have not yet been reported. We then investigated the role of steroid hormones in male mice using a skin flap ischemia model. APPROACH AND RESULTS: Castrated male mice developed skin necrosis after ischemia, whereas intact or castrated males treated with testosterone were equally protected. Testosterone can (1) activate the estrogen receptor after its aromatization into 17ß-estradiol or (2) be reduced into dihydrotestosterone, a nonaromatizable androgen that activates the androgen receptor. We found that dihydrotestosterone protected castrated wild-type mice by promoting skin revascularization, probably through a direct action on resistance arteries, as evidenced using a complementary model of flow-mediated outward remodeling. 17ß-estradiol treatment of castrated male mice also strongly protected them from ischemic necrosis through the activation of estrogen receptor-α by increasing skin revascularization and skin survival. Remarkably, 17ß-estradiol improved skin survival with a greater efficiency than dihydrotestosterone. CONCLUSIONS: Testosterone provides males with a strong protection against cutaneous necrosis and acts through both its estrogenic and androgenic derivatives, which have complementary effects on skin survival and revascularization.


Asunto(s)
Dihidrotestosterona/farmacología , Estradiol/farmacología , Terapia de Reemplazo de Hormonas , Isquemia/prevención & control , Neovascularización Fisiológica/efectos de los fármacos , Piel/irrigación sanguínea , Piel/efectos de los fármacos , Colgajos Quirúrgicos/irrigación sanguínea , Cicatrización de Heridas/efectos de los fármacos , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/genética , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Masculino , Arterias Mesentéricas/efectos de los fármacos , Ratones Pelados , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis , Orquiectomía , Ratas Wistar , Piel/metabolismo , Piel/patología , Colgajos Quirúrgicos/patología , Factores de Tiempo , Supervivencia Tisular/efectos de los fármacos
9.
Breast Cancer Res ; 18(1): 123, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27927249

RESUMEN

BACKGROUND: To date, all studies conducted on breast cancer diagnosis have focused on the expression of the full-length 66-kDa estrogen receptor alpha (ERα66). However, much less attention has been paid to a shorter 46-kDa isoform (ERα46), devoid of the N-terminal region containing the transactivation function AF-1. Here, we investigated the expression levels of ERα46 in breast tumors in relation to tumor grade and size, and examined the mechanism of its generation and its specificities of coregulatory binding and its functional activities. METHODS: Using approaches combining immunohistochemistry, Western blotting, and proteomics, antibodies allowing ERα46 detection were identified and the expression levels of ERα46 were quantified in 116 ERα-positive human breast tumors. ERα46 expression upon cellular stress was studied, and coregulator bindings, transcriptional, and proliferative response were determined to both ERα isoforms. RESULTS: ERα46 was expressed in over 70% of breast tumors at variable levels which sometimes were more abundant than ERα66, especially in differentiated, lower-grade, and smaller-sized tumors. We also found that ERα46 can be generated via internal ribosome entry site-mediated translation in the context of endoplasmic reticulum stress. The binding affinities of both unliganded and fully-activated receptors towards co-regulator peptides revealed that the respective potencies of ERα46 and ERα66 differ significantly, contributing to the differential transcriptional activity of target genes to 17ß estradiol (E2). Finally, increasing amounts of ERα46 decrease the proliferation rate of MCF7 tumor cells in response to E2. CONCLUSIONS: We found that, besides the full-length ERα66, the overlooked ERα46 isoform is also expressed in a majority of breast tumors. This finding highlights the importance of the choice of antibodies used for the diagnosis of breast cancer, which are able or not to detect the ERα46 isoform. In addition, since the function of both ERα isoforms differs, this work underlines the need to develop new technologies in order to discriminate ERα66 and ERα46 expression in breast cancer diagnosis which could have potential clinical relevance.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , Empalme Alternativo , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Análisis por Conglomerados , Estrés del Retículo Endoplásmico , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Unión Proteica , Biosíntesis de Proteínas , Isoformas de Proteínas , Proteoma , Proteómica/métodos , Estudios Retrospectivos
10.
Sci Rep ; 6: 33162, 2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27616470

RESUMEN

The mechanisms by which the airborne pathogen Mycobacterium tuberculosis spreads within the lung and leaves its primary niche to colonize other organs, thus inducing extrapulmonary forms of tuberculosis (TB) in humans, remains poorly understood. Herein, we used a transcriptomic approach to investigate the host cell gene expression profile in M. tuberculosis-infected human macrophages (ΜΦ). We identified 33 genes, encoding proteins involved in angiogenesis, for which the expression was significantly modified during infection, and we show that the potent angiogenic factor VEGF is secreted by M. tuberculosis-infected ΜΦ, in an RD1-dependent manner. In vivo these factors promote the formation of blood vessels in murine models of the disease. Inhibiting angiogenesis, via VEGF inactivation, abolished mycobacterial spread from the infection site. In accordance with our in vitro and in vivo results, we show that the level of VEGF in TB patients is elevated and that endothelial progenitor cells are mobilized from the bone marrow. These results strongly strengthen the most recent data suggesting that mycobacteria take advantage of the formation of new blood vessels to disseminate.


Asunto(s)
Pulmón/irrigación sanguínea , Mycobacterium tuberculosis/fisiología , Neovascularización Patológica/microbiología , Tuberculosis Pulmonar/fisiopatología , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Células Cultivadas , Femenino , Interacciones Huésped-Patógeno , Humanos , Pulmón/microbiología , Pulmón/patología , Ratones SCID , Neovascularización Patológica/metabolismo , Transcriptoma , Tuberculosis Pulmonar/microbiología , Regulación hacia Arriba
11.
Med Sci (Paris) ; 31(12): 1083-91, 2015 Dec.
Artículo en Francés | MEDLINE | ID: mdl-26672661

RESUMEN

Estrogen receptors (ER) are used as therapeutic targets, either for contraception or for the hormonal replacement therapy in menopausal women, but also in physiopathology for breast cancer treatment. It is therefore important to understand the tissue-specificity of the actions of ERα to optimize the benefits/risks ratio in each tissue. Besides the conventional nuclear ERα acting as a transcription factor, many studies have demonstrated that ERα is also able to mediate extra nuclear signaling, enabling rapid actions of estrogen. Recently, new transgenic mouse models were used to study these effects, and allowed to genetically segregate membrane versus nuclear actions of a steroid hormone receptor, demonstrating their in vivo tissue-specific roles.


Asunto(s)
Receptor alfa de Estrógeno/fisiología , Animales , Humanos , Especificidad de Órganos/fisiología
12.
Proc Natl Acad Sci U S A ; 111(2): E283-90, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24371309

RESUMEN

Estrogen receptor alpha (ERα) activation functions AF-1 and AF-2 classically mediate gene transcription in response to estradiol (E2). A fraction of ERα is targeted to plasma membrane and elicits membrane-initiated steroid signaling (MISS), but the physiological roles of MISS in vivo are poorly understood. We therefore generated a mouse with a point mutation of the palmitoylation site of ERα (C451A-ERα) to obtain membrane-specific loss of function of ERα. The abrogation of membrane localization of ERα in vivo was confirmed in primary hepatocytes, and it resulted in female infertility with abnormal ovaries lacking corpora lutea and increase in luteinizing hormone levels. In contrast, E2 action in the uterus was preserved in C451A-ERα mice and endometrial epithelial proliferation was similar to wild type. However, E2 vascular actions such as rapid dilatation, acceleration of endothelial repair, and endothelial NO synthase phosphorylation were abrogated in C451A-ERα mice. A complementary mutant mouse lacking the transactivation function AF-2 of ERα (ERα-AF2(0)) provided selective loss of function of nuclear ERα actions. In ERα-AF2(0), the acceleration of endothelial repair in response to estrogen-dendrimer conjugate, which is a membrane-selective ER ligand, was unaltered, demonstrating integrity of MISS actions. In genome-wide analysis of uterine gene expression, the vast majority of E2-dependent gene regulation was abrogated in ERα-AF2(0), whereas in C451A-ERα it was nearly fully preserved, indicating that membrane-to-nuclear receptor cross-talk in vivo is modest in the uterus. Thus, this work genetically segregated membrane versus nuclear actions of a steroid hormone receptor and demonstrated their in vivo tissue-specific roles.


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Receptor alfa de Estrógeno/genética , Ovario/fisiología , Útero/metabolismo , Análisis de Varianza , Animales , Western Blotting , Movimiento Celular , Biología Computacional , Células Endoteliales , Receptor alfa de Estrógeno/metabolismo , Femenino , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Lipoilación/genética , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Ovario/metabolismo , Mutación Puntual/genética , Receptor Cross-Talk/fisiología
13.
Cancer Res ; 72(12): 3010-9, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22523036

RESUMEN

Estrogens directly promote the growth of breast cancers that express the estrogen receptor α (ERα). However, the contribution of stromal expression of ERα in the tumor microenvironment to the protumoral effects of estrogen has never been explored. In this study, we evaluated the molecular and cellular mechanisms by which 17ß-estradiol (E2) impacts the microenvironment and modulates tumor development of ERα-negative tumors. Using different mouse models of ER-negative cancer cells grafted subcutaneously into syngeneic ovariectomized immunocompetent mice, we found that E2 potentiates tumor growth, increases intratumoral vessel density, and modifies tumor vasculature into a more regularly organized structure, thereby improving vessel stabilization to prevent tumor hypoxia and necrosis. These E2-induced effects were completely abrogated in ERα-deficient mice, showing a critical role of host ERα. Notably, E2 did not accelerate tumor growth when ERα was deficient in Tie2-positive cells, even in mice grafted with wild-type bone marrow. These results were extended by clinical evidence of ERα-positive stromal cell labeling in the microenvironment of human breast cancers. Together, our findings therefore show that E2 promotes the growth of ERα-negative cancer cells through the activation of stromal ERα (extra-hematopoietic Tie-2 positive cells), which normalizes tumor angiogenesis and allows an adaptation of blood supply to tumors, thereby preventing hypoxia and necrosis. These findings significantly deepen mechanistic insights into the impact of E2 on tumor development with potential consequences for cancer treatment.


Asunto(s)
Estradiol/metabolismo , Receptor alfa de Estrógeno/deficiencia , Melanoma/irrigación sanguínea , Melanoma/patología , Neovascularización Patológica/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Células del Estroma , Microambiente Tumoral
14.
Microbiology (Reading) ; 156(Pt 6): 1619-1631, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20185505

RESUMEN

Eukaryotic-like Ser/Thr protein kinases (STPKs) are present in many bacterial species, where they control various physiological and virulence processes by enabling microbial adaptation to specific environmental signals. PknJ is the only member of the 11 STPKs identified in Mycobacterium tuberculosis that still awaits characterization. Here we report that PknJ is a functional kinase that forms dimers in vitro, and contains a single transmembrane domain. Using a high-density peptide-chip-based technology, multiple potential mycobacterial targets were identified for PknJ. We confirmed PknJ-dependent phosphorylation of four of these targets: PknJ itself, which autophosphorylates at Thr(168), Thr(171) and Thr(173) residues; the transcriptional regulator EmbR; the methyltransferase MmaA4/Hma involved in mycolic acid biosynthesis; and the dipeptidase PepE, whose encoding gene is located next to pknJ in the mycobacterial genome. Our results provide a number of candidate phospho-targets for PknJ and possibly other mycobacterial STPKs that could be studied to investigate the role of STPKs in M. tuberculosis physiology and virulence.


Asunto(s)
Mycobacterium tuberculosis/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Dimerización , Ratones , Datos de Secuencia Molecular , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Alineación de Secuencia , Serina/metabolismo , Transducción de Señal , Treonina/metabolismo , Tuberculosis/microbiología , Virulencia
15.
J Exp Med ; 206(10): 2205-20, 2009 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-19770268

RESUMEN

The C-type lectin dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN) mediates the innate immune recognition of microbial carbohydrates. We investigated the function of this molecule in the host response to pathogens in vivo, by generating mouse lines lacking the DC-SIGN homologues SIGNR1, SIGNR3, and SIGNR5. Resistance to Mycobacterium tuberculosis was impaired only in SIGNR3-deficient animals. SIGNR3 was expressed in lung phagocytes during infection, and interacted with M. tuberculosis bacilli and mycobacterial surface glycoconjugates to induce secretion of critical host defense inflammatory cytokines, including tumor necrosis factor (TNF). SIGNR3 signaling was dependent on an intracellular tyrosine-based motif and the tyrosine kinase Syk. Thus, the mouse DC-SIGN homologue SIGNR3 makes a unique contribution to protection of the host against a pulmonary bacterial pathogen.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Lectinas Tipo C/fisiología , Receptores de Superficie Celular/fisiología , Tuberculosis/inmunología , Animales , Antígenos CD/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Glicoconjugados/metabolismo , Interleucina-6/biosíntesis , Lipopolisacáridos/metabolismo , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Transducción de Señal , Receptor Toll-Like 2/fisiología , Factor de Necrosis Tumoral alfa/biosíntesis
16.
Vaccine ; 27(1): 28-37, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18977269

RESUMEN

The potent IFN-gamma inducing fusion antigen Ag85B-ESAT-6 (85B6) is a lead subunit candidate to improve current vaccination against Mycobacterium tuberculosis (Mtb). The recombinant M. bovis BCG strain Myc3504 was constructed to secrete 85B6. It was based on commercial BCG strain Moreau Rio de Janeiro (BCG(MoWT)) which remains available for human oral administration. Myc 3504 induced higher levels of 85B6-specific IFN-gamma circulating T-cells as compared to BCG(MoWT). A novel needle-free mucosal immunization regimen combining oral prime with Myc3504 or BCG(MoWT) with intranasal boost with LTK-63-adjuvanted 85B6 was compared to subcutaneous prime-boost immunization. Strikingly whereas parenteral immunization induced sustained levels of 85B6-specific IFN-gamma secretion by circulating T-cells, mucosal regimens induced barely detectable IFN-gamma. Despite this, mice and guinea pigs immunized with the mucosal regimens were as efficiently protected against aerosol Mtb challenge as parenterally immunized animals. After Mtb challenge, anti-ESAT-6 IFN-gamma responses sharply increased in non-vaccinated mice as a hallmark of infection. Parenterally immunized mice that controlled Mtb infection, displayed anti-ESAT-6 IFN-gamma responses as high as non-immunized infected mice, compromising the possible use of ESAT-6 as a diagnostic tool. Interestingly, in mucosally immunized mice that were equally protected, post-challenge ESAT-6-specific IFN-gamma T-cell response remained low.


Asunto(s)
Aciltransferasas/inmunología , Antígenos Bacterianos/inmunología , Vacuna BCG , Proteínas Bacterianas/inmunología , Interferón gamma/inmunología , Linfocitos T/inmunología , Tuberculosis/prevención & control , Administración Intranasal , Animales , Vacuna BCG/administración & dosificación , Vacuna BCG/química , Vacuna BCG/inmunología , Femenino , Cobayas , Humanos , Inmunización Secundaria , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Tuberculosis/inmunología
17.
Microbes Infect ; 10(12-13): 1379-86, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18771745

RESUMEN

We recently showed that treatment of macrophages prior to Mycobacterium tuberculosis infection with the pro-inflammatory omega-6 lipid, arachidonic acid (AA) enhanced bacterial killing whereas the anti-inflammatory, omega-3 lipid eicosapentaenoic acid (EPA) stimulated bacterial growth. Here we tested if these effects were depending on when lipids were added to macrophages: before or during Mycobacterium smegmatis or M. tuberculosis infection. Collectively, our data suggested that a high omega-6 diet might be beneficial against mycobacteriosis, while a high omega-3 diet might be detrimental. AA also stimulated TNF-alpha secretion in M. tuberculosis-infected macrophages whereas EPA inhibited this process. AA strongly activated the MAP kinase p38 in uninfected cells but M. tuberculosis infected cells blocked the ability of AA to activate p38; AA-dependent killing is therefore independent of p38. We therefore tested diets enriched in omega-3 and omega-6 lipids on a mouse model of tuberculosis. In contrast to the in vitro results, the omega-6 tended to increase survival of M. tuberculosis in mice, while omega-3- tended to increase pathogen killing. Overall our results together with those previously reported in the literature suggest that it is almost impossible to predict, at the whole organism level, if a diet enriched in omega-3 or -6 will be beneficial or detrimental to intracellular pathogens.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-6/farmacología , Interacciones Huésped-Patógeno , Macrófagos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/fisiología , Animales , Ácido Araquidónico/administración & dosificación , Ácido Araquidónico/farmacología , Línea Celular , Células Cultivadas , Ácido Eicosapentaenoico/administración & dosificación , Ácido Eicosapentaenoico/farmacología , Ácidos Grasos Omega-3/administración & dosificación , Ácidos Grasos Omega-6/administración & dosificación , Femenino , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/crecimiento & desarrollo , Mycobacterium tuberculosis/crecimiento & desarrollo , Tuberculosis/inmunología , Tuberculosis/microbiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
J Bacteriol ; 190(4): 1329-34, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18065542

RESUMEN

Similarities between Mycobacterium tuberculosis phoP-phoR mutants and the attenuated laboratory strain M. tuberculosis H37Ra in terms of morphological and cytochemical properties, lipid content, gene expression and virulence attenuation prompted us to analyze the functionality of this two-component regulator in the latter strain. Sequence analysis revealed a base substitution resulting in a one-amino-acid change in the likely DNA-binding region of PhoP in H37Ra relative to H37Rv. Using gel-shift assays, we show that this mutation abrogates the ability of the H37Ra PhoP protein to bind to a 40-bp segment of its own promoter. Consistent with this result, the phoP gene from H37Rv but not that from H37Ra was able to restore the synthesis of sulfolipids, diacyltrehaloses and polyacyltrehaloses in an isogenic phoP-phoR knock-out mutant of M. tuberculosis Moreover, complementation of H37Ra with phoP from H37Rv fully restored sulfolipid, diacyltrehalose and polyacyltrehalose synthesis, clearly indicating that the lack of production of these lipids in H37Ra is solely due to the point mutation in phoP. Using a pks2-3/4 knock-out mutant of M. tuberculosis H37Rv, evidence is further provided that the above-mentioned polyketide-derived acyltrehaloses do not significantly contribute to the virulence of the tubercle bacillus in a mouse model of infection. Reasons for the attenuation of H37Ra thus most likely stand in other virulence factors, many of which are expected to belong to the PhoP regulon and another of which, unrelated to PhoP, appears to be the lack of production of phthiocerol dimycocerosates in this strain.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , Mutación Puntual , Trehalosa/metabolismo , Animales , Proteínas Bacterianas/genética , Dicroismo Circular , Ensayo de Cambio de Movilidad Electroforética , Eliminación de Gen , Prueba de Complementación Genética , Glucolípidos/química , Glucolípidos/metabolismo , Lípidos/química , Lípidos/genética , Macrólidos/metabolismo , Ratones , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Recombinantes/metabolismo , Trehalosa/química , Tuberculosis/microbiología , Virulencia/genética
19.
J Biol Chem ; 282(37): 27270-27276, 2007 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-17640872

RESUMEN

Mycobacteria produce two unusual polymethylated polysaccharides, the 6-O-methylglucosyl-containing lipopolysaccharides (MGLP) and the 3-O-methylmannose polysaccharides, which have been shown to regulate fatty acid biosynthesis in vitro. A cluster of genes dedicated to the synthesis of MGLP was identified in Mycobacterium tuberculosis and Mycobacterium smegmatis. Overexpression of the putative glycosyltransferase gene Rv3032 in M. smegmatis greatly stimulated MGLP production, whereas the targeted disruption of Rv3032 in M. tuberculosis and that of the putative methyltransferase gene MSMEG2349 in M. smegmatis resulted in a dramatic reduction in the amounts of MGLP synthesized and in the accumulation of precursors of these molecules. Disruption of Rv3032 also led to a significant decrease in the glycogen content of the tubercle bacillus, indicating that the product of this gene is likely to be involved in the elongation of more than one alpha-(1-->4)-glucan in this bacterium. Results thus suggest that Rv3032 encodes the alpha-(1-->4)-glucosyltransferase responsible for the elongation of MGLP, whereas MSMEG2349 encodes the O-methyltransferase required for the 6-O-methylation of these compounds.


Asunto(s)
Lipopolisacáridos/biosíntesis , Metilglucósidos/análisis , Mycobacterium tuberculosis/metabolismo , Glucógeno/análisis , Glicosiltransferasas/genética , Glicosiltransferasas/fisiología , Familia de Multigenes , Mycobacterium tuberculosis/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Infect Immun ; 75(1): 504-7, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17030567

RESUMEN

Using signature-tagged transposon mutagenesis, we isolated 23 Mycobacterium tuberculosis mutants, corresponding to 21 genes or genetic regions, attenuated in their ability to parasitize human macrophages. Mutants disrupted in the ABC transporter-encoding genes Rv0986 and Rv0987 were further characterized as being impaired in their ability to bind to host cells.


Asunto(s)
Elementos Transponibles de ADN/genética , Genes Bacterianos , Macrófagos/parasitología , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Animales , Proteínas Bacterianas/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Mutagénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA