Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33771918

RESUMEN

Knockout of ERß in the mouse leads to nuclear expression of epidermal growth factor receptor (EGFR) in the prostate. To examine whether ERß plays a similar role in the human prostate, we used four cohorts of men: 1) a Swedish cohort of normal prostates and PCa (prostate cancer) of different Gleason grades; 2) men with benign prostatic hyperplasia (BPH) treated with the 5α-reductase inhibitor, finasteride, and finasteride together with the ERß agonists, soy isoflavones; 3) men with PCa above Gleason grade 4 (GG4), treated with ADT (androgen deprivation therapy) and abiraterone (AA), the blocker of androgen synthesis for different durations; and 4) men with GG4 PCa on ADT or ADT with the AR (androgen receptor) blocker, enzalutamide, for 4 mo to 6 mo. In men with BPH, finasteride treatment induced EGFR nuclear expression, but, when finasteride was combined with isoflavones, EGFR remained on the cell membrane. In GG4 patients, blocking of AR for 4 mo to 6 mo resulted in loss of ERß and PTEN expression and increase in patients with nuclear EGFR from 10 to 40%. In the men with GG4 PCa, blocking of adrenal synthesis of testosterone for 2 mo to 7 mo had the beneficial effect of increasing ERß expression, but, on treatment longer than 8 mo, ERß was lost and EGFR moved to the nucleus. Since nuclear EGFR is a predictor of poor outcome in PCa, addition of ERß agonists together with abiraterone should be considered as a treatment that might sustain expression of ERß and offer some benefit to patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Receptor beta de Estrógeno/agonistas , Hiperplasia Prostática/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Transporte Activo de Núcleo Celular/efectos de los fármacos , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Androstenos/farmacología , Androstenos/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzamidas/farmacología , Benzamidas/uso terapéutico , Biopsia , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Estudios de Cohortes , Receptores ErbB/metabolismo , Receptor beta de Estrógeno/metabolismo , Finasterida/farmacología , Finasterida/uso terapéutico , Humanos , Masculino , Ratones , Ratones Noqueados , Clasificación del Tumor , Nitrilos/farmacología , Nitrilos/uso terapéutico , Fosfohidrolasa PTEN/metabolismo , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Fitoestrógenos/farmacología , Fitoestrógenos/uso terapéutico , Próstata/citología , Próstata/efectos de los fármacos , Próstata/patología , Hiperplasia Prostática/patología , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34389675

RESUMEN

To identify regulators of triple-negative breast cancer (TNBC), gene expression profiles of malignant parts of TNBC (mTNBC) and normal adjacent (nadj) parts of the same breasts have been compared. We are interested in the roles of estrogen receptor ß (ERß) and the cytochrome P450 family (CYPs) as drivers of TNBC. We examined by RNA sequencing the mTNBC and nadj parts of five women. We found more than a fivefold elevation in mTNBC of genes already known to be expressed in TNBC: BIRC5/survivin, Wnt-10A and -7B, matrix metalloproteinases (MMPs), chemokines, anterior gradient proteins, and lysophosphatidic acid receptor and the known basal characteristics of TNBC, sox10, ROPN1B, and Col9a3. There were two unexpected findings: 1) a strong induction of CYPs involved in activation of fatty acids (CYP4), and in inactivation of calcitriol (CYP24A1) and retinoic acid (CYP26A1); and 2) a marked down-regulation of FOS, FRA1, and JUN, known tethering partners of ERß. ERß is expressed in 20 to 30% of TNBCs and is being evaluated as a target for treating TNBC. We used ERß+ TNBC patient-derived xenografts in mice and found that the ERß agonist LY500703 had no effect on growth or proliferation. Expression of CYPs was confirmed by immunohistochemistry in formalin-fixed and paraffin-embedded (FFPE) TNBC. In TNBC cell lines, the CYP4Z1-catalyzed fatty acid metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) increased proliferation, while calcitriol decreased proliferation but only after inhibition of CYP24A1. We conclude that CYP-mediated pathways can be drivers of TNBC but that ERß is unlikely to be a tumor suppressor because the absence of its main tethering partners renders ERß functionless on genes involved in proliferation and inflammation.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Receptor beta de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Anfibias/genética , Proteínas Anfibias/metabolismo , Animales , Benzopiranos/farmacología , Calcitriol/farmacología , Sistema Enzimático del Citocromo P-450/genética , Regulación hacia Abajo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Ácidos Grasos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Ratones , Neoplasias Experimentales , Distribución Aleatoria , Survivin/genética , Survivin/metabolismo , Transcriptoma , Tretinoina/farmacología , Neoplasias de la Mama Triple Negativas/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(42): 26347-26355, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33020300

RESUMEN

Loss of the tumor suppressor, PTEN, is one of the most common findings in prostate cancer (PCa). This loss leads to overactive Akt signaling, which is correlated with increased metastasis and androgen independence. However, another tumor suppressor, inositol-polyphosphate 4-phosphatase type II (INPP4B), can partially compensate for the loss of PTEN. INPP4B is up-regulated by androgens, and this suggests that androgen-deprivation therapy (ADT) would lead to hyperactivity of AKT. However, in the present study, we found that in PCa, samples from men treated with ADT, ERß, and INPP4B expression were maintained in some samples. To investigate the role of ERß1 in regulation of INPPB, we engineered the highly metastatic PCa cell line, PC3, to express ERß1. In these cells, INPP4B was induced by ERß ligands, and this induction was accompanied by inhibition of Akt activity and reduction in cell migration. These findings reveal that, in the absence of androgens, ERß1 induces INPP4B to dampen AKT signaling. Since the endogenous ERß ligand, 3ß-Adiol, is lost upon long-term ADT, to obtain the beneficial effects of ERß1 on AKT signaling, an ERß agonist should be added along with ADT.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antagonistas de Andrógenos/metabolismo , Andrógenos/farmacología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Humanos , Masculino , Células PC-3 , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal
4.
Proc Natl Acad Sci U S A ; 117(9): 4902-4909, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32075916

RESUMEN

Disagreements about the phenotype of estrogen receptor ß (ERß) knockout mouse, created by removing the DNA-binding domain of the ERß gene or interruption of the gene with a neocassette (Oliver Smithies ERß knockout mice [ERßOS-/-]), prompted us to create an ERß knockout mouse by deleting the ERß gene with the use of CRISPR/Cas9 technology. We confirmed that the ERß gene was eliminated from the mouse genome and that no ERß mRNA or protein was detectable in tissues of this mouse. Overall the phenotype of the ventral prostate (VP) and mammary gland (MG) in ERßcrispr-/- mice was similar to, but more severe than, that in the ERßOS-/-mice. In the VP of 6-mo-old ERßcrispr-/- mice there was epithelial hyperplasia, fibroplasia, inflammation, stromal overgrowth, and intraductal cancer-like lesions. This was accompanied by an increase in Ki67 and P63 and loss in DACH1 and PURα, two androgen receptor (AR) repressors. In the MG there was overexpression of estrogen receptor α and progesterone receptor, loss of collagen, increase in proliferation and expression of metalloproteases, and invasive epithelium. Surprisingly, by 18 mo of age, the number of hyperplastic foci was reduced, the ducts of the VP and MG became atrophic, and, in the VP, there was massive immune infiltration and massive desquamation of the luminal epithelial cells. These changes were coincident with reduced levels of androgens in males and estrogens in females. We conclude that ERß is a tumor suppressor gene in the VP and MG where its loss increases the activity AR and ERα, respectively.


Asunto(s)
Receptor beta de Estrógeno/genética , Glándulas Mamarias Animales/metabolismo , Fenotipo , Próstata/metabolismo , Receptores Androgénicos/metabolismo , Eliminación de Secuencia , Andrógenos/metabolismo , Animales , Sistemas CRISPR-Cas , Quimiocinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Epitelio/metabolismo , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Proteínas del Ojo , Femenino , Hiperplasia/patología , Inflamación , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Próstata/patología , ARN Mensajero/metabolismo , Transducción de Señal , Células del Estroma , Transactivadores , Factores de Transcripción/metabolismo , Transcriptoma , Proteínas Supresoras de Tumor/metabolismo
5.
Int J Mol Sci ; 24(15)2023 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-37569842

RESUMEN

Liver X receptors (LXRα and LXRß) are oxysterol-activated nuclear receptors that play key roles in cholesterol homeostasis, the central nervous system, and the immune system. We have previously reported that LXRαß-deficient mice are more susceptible to dextran sodium sulfate (DSS)-induced colitis than their WT littermates, and that an LXR agonist protects against colitis in mice mainly via the regulation of the immune system in the gut. We now report that both LXRα and LXRß are expressed in the colonic epithelium and that in aging LXRαß-/- mice there is a reduction in the intensity of goblet cells, mucin (MUC2), TFF3, and estrogen receptor ß (ERß) levels. The cytoplasmic compartment of the surface epithelial cells was markedly reduced and there was a massive invasion of macrophages in the lamina propria. The expression and localization of ß-catenin, α-catenin, and E-cadherin were not changed, but the shrinkage of the cytoplasm led to an appearance of an increase in staining. In the colonic epithelium there was a reduction in the expression of plectin, a hemidesmosome protein whose loss in mice leads to spontaneous colitis, ELOVL1, a fatty acid elongase protein coding gene whose overexpression is found in colorectal cancer, and non-neuronal choline acetyltransferase (ChAT) involved in the regulation of epithelial cell adhesion. We conclude that in aging LXRαß-/- mice, the phenotype in the colon is due to loss of ERß expression.


Asunto(s)
Colitis , Receptor beta de Estrógeno , Ratones , Animales , Receptor beta de Estrógeno/metabolismo , Ratones Noqueados , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Colon/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Mucosa Intestinal/metabolismo , Sulfato de Dextran/toxicidad , Ratones Endogámicos C57BL
6.
Proc Natl Acad Sci U S A ; 116(33): 16507-16512, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31371497

RESUMEN

The retina is an extension of the brain. Like the brain, neurodegeneration of the retina occurs with age and is the cause of several retinal diseases including optic neuritis, macular degeneration, and glaucoma. Liver X receptors (LXRs) are expressed in the brain where they play a key role in maintenance of cerebrospinal fluid and the health of dopaminergic neurons. Herein, we report that LXRs are expressed in the retina and optic nerve and that loss of LXRß, but not LXRα, leads to loss of ganglion cells in the retina. In the retina of LXRß-/- mice, there is an increase in amyloid A4 and deposition of beta-amyloid (Aß) aggregates but no change in the level of apoptosis or autophagy in the ganglion cells and no activation of microglia or astrocytes. However, in the optic nerve there is a loss of aquaporin 4 (AQP4) in astrocytes and an increase in activation of microglia. Since loss of AQP4 and microglial activation in the optic nerve precedes the loss of ganglion cells, and accumulation of Aß in the retina, the cause of the neuronal loss appears to be optic nerve degeneration. In patients with optic neuritis there are frequently AQP4 autoantibodies which block the function of AQP4. LXRß-/- mouse is another model of optic neuritis in which AQP4 antibodies are not detectable, but AQP4 function is lost because of reduction in its expression.


Asunto(s)
Receptores X del Hígado/deficiencia , Degeneración Nerviosa/patología , Nervio Óptico/patología , Retina/patología , Péptidos beta-Amiloides/metabolismo , Animales , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Femenino , Receptores X del Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Oligodendroglía/metabolismo , Nervio Óptico/metabolismo , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología
7.
Neuroendocrinology ; 111(1-2): 27-44, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-31991411

RESUMEN

BACKGROUND: Male estrogen receptor beta (ERß) knockout (BERKO) mice display anxiety and aggression linked to, among others, altered serotonergic signaling in the basolateral amygdala and dorsal raphe, impaired cortical radial glia migration, and reduced GABAergic signaling. The effects on primary motor cortex (M1 cortex) and locomotor activity as a consequence of ERß loss have not been investigated. OBJECTIVE: The aim of this study was to determine whether locomotor activity is altered as a consequence of the changes in the M1 cortex. METHODS: The locomotor activity of male wild-type (WT) and BERKO mice was evaluated using the open-field and rotarod tests. Molecular changes in the M1 cortex were analyzed by RNA sequencing, electron microscopy, electrophysiology, and immunohistological techniques. In addition, we established oligodendrocyte (OL) cultures from WT and BERKO mouse embryonic stem cells to evaluate OL function. RESULTS: Locomotor profiling revealed that BERKO mice were more active than WT mice but had impaired motor coordination. Analysis of the M1 cortex pointed out differences in synapse function and myelination. There was a reduction in GABAergic signaling resulting in imbalanced excitatory and inhibitory neurotransmission as well as a defective OL differentiation accompanied by myelin defects. The effects of ERß loss on OL differentiation were confirmed in vitro. CONCLUSION: ERß is an important regulator of GABAergic interneurons and OL differentiation, which impacts on adult M1 cortex function and may be linked to increased locomotor activity and decreased motor coordination in BERKO mice.


Asunto(s)
Receptor beta de Estrógeno/genética , Locomoción/genética , Corteza Motora/fisiopatología , Vaina de Mielina/fisiología , Desempeño Psicomotor , Transmisión Sináptica , Animales , Expresión Génica , Masculino , Ratones , Ratones Noqueados , Corteza Motora/metabolismo , Oligodendroglía/patología
8.
Proc Natl Acad Sci U S A ; 114(19): E3816-E3822, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28439009

RESUMEN

As estrogen receptor ß-/- (ERß-/-) mice age, the ventral prostate (VP) develops increased numbers of hyperplastic, fibroplastic lesions and inflammatory cells. To identify genes involved in these changes, we used RNA sequencing and immunohistochemistry to compare gene expression profiles in the VP of young (2-mo-old) and aging (18-mo-old) ERß-/- mice and their WT littermates. We also treated young and old WT mice with an ERß-selective agonist and evaluated protein expression. The most significant findings were that ERß down-regulates androgen receptor (AR) signaling and up-regulates the tumor suppressor phosphatase and tensin homolog (PTEN). ERß agonist increased expression of the AR corepressor dachshund family (DACH1/2), T-cadherin, stromal caveolin-1, and nuclear PTEN and decreased expression of RAR-related orphan receptor c, Bcl2, inducible nitric oxide synthase, and IL-6. In the ERß-/- mouse VP, RNA sequencing revealed that the following genes were up-regulated more than fivefold: Bcl2, clusterin, the cytokines CXCL16 and -17, and a marker of basal/intermediate cells (prostate stem cell antigen) and cytokeratins 4, 5, and 17. The most down-regulated genes were the following: the antioxidant gene glutathione peroxidase 3; protease inhibitors WAP four-disulfide core domain 3 (WFDC3); the tumor-suppressive genes T-cadherin and caveolin-1; the regulator of transforming growth factor ß signaling SMAD7; and the PTEN ubiquitin ligase NEDD4. The role of ERß in opposing AR signaling, proliferation, and inflammation suggests that ERß-selective agonists may be used to prevent progression of prostate cancer, prevent fibrosis and development of benign prostatic hyperplasia, and treat prostatitis.


Asunto(s)
Envejecimiento/metabolismo , Regulación hacia Abajo , Receptor beta de Estrógeno/metabolismo , Próstata/metabolismo , Receptores Androgénicos/biosíntesis , Transducción de Señal , Envejecimiento/genética , Envejecimiento/patología , Andrógenos/metabolismo , Animales , Quimiocina CXCL16/biosíntesis , Quimiocina CXCL16/genética , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/genética , Clusterina/biosíntesis , Clusterina/genética , Receptor beta de Estrógeno/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Queratinas/biosíntesis , Queratinas/genética , Masculino , Ratones , Ratones Noqueados , Ubiquitina-Proteína Ligasas Nedd4/biosíntesis , Ubiquitina-Proteína Ligasas Nedd4/genética , Fosfohidrolasa PTEN/biosíntesis , Fosfohidrolasa PTEN/genética , Próstata/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores Androgénicos/genética , Proteína smad7/biosíntesis , Proteína smad7/genética
9.
Proc Natl Acad Sci U S A ; 113(42): 11883-11888, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27688768

RESUMEN

The aryl hydrocarbon receptor (AhR) is now recognized as an important physiological regulator in the immune and reproductive systems, and in the development of the liver and vascular system. AhR regulates cell cycle, cell proliferation, and differentiation through interacting with other signaling pathways, like estrogen receptor α (ERα), androgen receptor (AR), and Notch signaling. In the present study, we investigated Notch and estrogen signaling in AhR-/- mice. We found low fertility with degenerative changes in the testes, germ cell apoptosis, and a reduced number of early spermatids. There was no change in aromatase, AR, ERα, or ERß expression in the testis and no detectable change in serum estrogen levels. However, expression of Notch receptors (Notch1 and Notch3) and their target Hairy and Enhancer of Split homolog 1 (HES1) was reduced. In addition, the testosterone level was slightly reduced in the serum. In the mammary fat pad, AhR appeared to regulate estrogen signaling because, in AhR-/- males, there was significant growth of the mammary ducts with high expression of ERα in the ductal epithelium. The enhanced mammary ductal growth appears to be related to overexpression of ERα accompanied by a high proliferation index, whereas the reduced fertility appears to be related defects in Notch signaling that leads to reduced expression of HES1 and, consequently, early maturation of spermatocytes and a depletion of primary spermatids. Previous reports have indicated that AhR pathway is associated with infertility in men. Our results provide a mechanistic explanation for this defect.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Receptor alfa de Estrógeno/metabolismo , Receptores de Hidrocarburo de Aril/deficiencia , Receptores Notch/metabolismo , Transducción de Señal , Animales , Apoptosis/genética , Aromatasa/metabolismo , Biomarcadores , Proliferación Celular , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Fertilidad/genética , Eliminación de Gen , Expresión Génica , Células Germinativas/metabolismo , Inmunohistoquímica , Masculino , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Espermatocitos/metabolismo , Testículo/metabolismo
10.
Proc Natl Acad Sci U S A ; 113(27): 7614-9, 2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27335465

RESUMEN

The etiology of peripheral squamous cell lung cancer (PSCCa) remains unknown. Here, we show that this condition spontaneously develops in mice in which the genes for two oxysterol receptors, Liver X Receptor (LXR) α (Nr1h3) and ß (Nr1h2), are inactivated. By 1 y of age, most of these mice have to be euthanized because of severe dyspnea. Starting at 3 mo, the lungs of LXRα,ß(Dko) mice, but not of LXRα or LXRß single knockout mice, progressively accumulate foam cells, so that by 1 y, the lungs are covered by a "golden coat." There is infiltration of inflammatory cells and progressive accumulation of lipid in the alveolar wall, type 2 pneumocytes, and macrophages. By 14 mo, there are three histological lesions: one resembling adenomatous hyperplasia, one squamous metaplasia, and one squamous cell carcinoma characterized by expression of transformation-related protein (p63), sex determining region Y-box 2 (Sox2), cytokeratin 14 (CK14), and cytokeratin 13 (CK13) and absence of thyroid transcription factor 1 (TTF1), and prosurfactant protein C (pro-SPC). RNA sequencing analysis at 12 mo confirmed a massive increase in markers of M1 macrophages and lymphocytes. The data suggest a previously unidentified etiology of PSCCa: cholesterol dysregulation and M1 macrophage-predominant lung inflammation combined with damage to, and aberrant repair of, lung tissue, particularly the peripheral parenchyma. The results raise the possibility that components of the LXR signaling may be useful targets in the treatment of PSCCa.


Asunto(s)
Metabolismo de los Lípidos , Receptores X del Hígado/fisiología , Neoplasias Pulmonares/etiología , Pulmón/metabolismo , Neoplasias de Células Escamosas/etiología , Células Epiteliales Alveolares/metabolismo , Animales , Fibroblastos/metabolismo , Homeostasis , Pulmón/patología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Neumonía/etiología , Análisis de Secuencia de ARN
11.
Proc Natl Acad Sci U S A ; 112(45): 14006-11, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26504234

RESUMEN

The recent discovery of browning of white adipose tissue (WAT) has raised great research interest because of its significant potential in counteracting obesity and type 2 diabetes. Browning is the result of the induction in WAT of a newly discovered type of adipocyte, the beige cell. When mice are exposed to cold or several kinds of hormones or treatments with chemicals, specific depots of WAT undergo a browning process, characterized by highly activated mitochondria and increased heat production and energy expenditure. However, the mechanisms underlying browning are still poorly understood. Liver X receptors (LXRs) are one class of nuclear receptors, which play a vital role in regulating cholesterol, triglyceride, and glucose metabolism. Following our previous finding that LXRs serve as repressors of uncoupling protein-1 (UCP1) in classic brown adipose tissue in female mice, we found that LXRs, especially LXRß, also repress the browning process of subcutaneous adipose tissue (SAT) in male rodents fed a normal diet. Depletion of LXRs activated thyroid-stimulating hormone (TSH)-releasing hormone (TRH)-positive neurons in the paraventricular nucleus area of the hypothalamus and thus stimulated secretion of TSH from the pituitary. Consequently, production of thyroid hormones in the thyroid gland and circulating thyroid hormone level were increased. Moreover, the activity of thyroid signaling in SAT was markedly increased. Together, our findings have uncovered the basis of increased energy expenditure in male LXR knockout mice and provided support for targeting LXRs in treatment of obesity.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Encéfalo/metabolismo , Metabolismo Energético/fisiología , Mitocondrias/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Hormonas Tiroideas/metabolismo , Análisis de Varianza , Animales , Composición Corporal/fisiología , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Inmunohistoquímica , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos/genética , Hormona Liberadora de Tirotropina/metabolismo
12.
Breast Cancer Res ; 19(1): 79, 2017 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-28673316

RESUMEN

BACKGROUND: Upregulation of estrogen receptor beta (ERß) in breast cancer cells is associated with epithelial maintenance, decreased proliferation and invasion, and a reduction in the expression of the receptor has been observed in invasive breast tumors. However, proof of an association between loss of ERß and breast carcinogenesis is still missing. METHODS: To study the role of ERß in breast oncogenesis, we generated mouse conditional mutants with specific inactivation of ERß and p53 in the mammary gland epithelium. For epithelium-specific knockout of ERß and p53, ERß F/F and p53 F/F mice were crossed to transgenic mice that express the Cre recombinase under the control of the human keratin 14 promoter. RESULTS: Somatic loss of ERß significantly accelerated formation of p53-deficient mammary tumors. Loss of the receptor also resulted in the development of less differentiated carcinomas with stronger spindle cell morphology and decreased expression of luminal epithelial markers. CONCLUSIONS: Our results show that synergism between ERß and p53 inactivation functions to determine important aspects of breast oncogenesis and cancer progression.


Asunto(s)
Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Receptor beta de Estrógeno/genética , Proteína p53 Supresora de Tumor/genética , Alelos , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Transición Epitelial-Mesenquimal/genética , Epitelio/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Expresión Génica , Silenciador del Gen , Genotipo , Inmunohistoquímica , Estimación de Kaplan-Meier , Ratones , Ratones Transgénicos , Fenotipo , Pronóstico , Proteína p53 Supresora de Tumor/metabolismo
13.
Proc Natl Acad Sci U S A ; 111(6): 2277-82, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24464484

RESUMEN

The farnesoid X receptor (FXR) is a ligand-activated transcription factor belonging to the nuclear receptor superfamily. FXR is mainly expressed in liver and small intestine, where it plays an important role in bile acid, lipid, and glucose metabolism. The kidney also has a high FXR expression level, with its physiological function unknown. Here we demonstrate that FXR is ubiquitously distributed in renal tubules. FXR agonist treatment significantly lowered urine volume and increased urine osmolality, whereas FXR knockout mice exhibited an impaired urine concentrating ability, which led to a polyuria phenotype. We further found that treatment of C57BL/6 mice with chenodeoxycholic acid, an FXR endogenous ligand, significantly up-regulated renal aquaporin 2 (AQP2) expression, whereas FXR gene deficiency markedly reduced AQP2 expression levels in the kidney. In vitro studies showed that the AQP2 gene promoter contained a putative FXR response element site, which can be bound and activated by FXR, resulting in a significant increase of AQP2 transcription in cultured primary inner medullary collecting duct cells. In conclusion, the present study demonstrates that FXR plays a critical role in the regulation of urine volume, and its activation increases urinary concentrating capacity mainly via up-regulating its target gene AQP2 expression in the collecting ducts.


Asunto(s)
Capacidad de Concentración Renal/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Secuencia de Bases , Cartilla de ADN , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Receptores Citoplasmáticos y Nucleares/metabolismo
14.
Proc Natl Acad Sci U S A ; 110(9): 3543-8, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23401502

RESUMEN

A therapeutic goal in the treatment of certain CNS diseases, including multiple sclerosis, amyotrophic lateral sclerosis, and Parkinson disease, is to down-regulate inflammatory pathways. Inflammatory molecules produced by microglia are responsible for removal of damaged neurons, but can cause collateral damage to normal neurons located close to defective neurons. Although estrogen can inactivate microglia and inhibit the recruitment of T cells and macrophages into the CNS, there is controversy regarding which of the two estrogen receptors (ERs), ERα or ERß, mediates the beneficial effects in microglia. In this study, we found that ERß, but not ERα, is expressed in microglia. Using the experimental autoimmune encephalomyelitis (EAE) model in SJL/J mice, we evaluated the benefit of an ERß agonist as a modulator of neuroinflammation. Treatment of EAE mice with LY3201, a selective ERß agonist provided by Eli Lilly, resulted in marked reduction of activated microglia in the spinal cord. LY3201 down-regulated the nuclear transcription factor NF-κB, as well as the NF-κB-induced gene inducible nitric oxide synthase in microglia and CD3(+) T cells. In addition, LY3201 inhibited T-cell reactivity through regulation of indoleamine-2,3-dioxygenase. In the EAE model, treatment with LY3201 decreased mortality in the first 2 wk after disease onset, and also reduced the severity of symptoms in mice surviving for 4 wk. Our data show that ERß-selective agonists, by modulating the immune system in both microglia and T cells, offer promise as a useful class of drugs for treating degenerative diseases of the CNS.


Asunto(s)
Benzopiranos/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Receptor beta de Estrógeno/metabolismo , Microglía/metabolismo , Terapia Molecular Dirigida , Linfocitos T/metabolismo , Animales , Benzopiranos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/patología , Receptor alfa de Estrógeno/metabolismo , Femenino , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/patología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/enzimología , Médula Espinal/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología
15.
Cell Physiol Biochem ; 37(1): 67-76, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26278717

RESUMEN

BACKGROUND/AIMS: HIF-1α is accumulated in the cellular nucleus and cytoplasm under conditions of oxygen deprivation and engaged in pathophysiologic changes of homeostasis by modulating the expression of several target genes. As an endogenous signaling protein, HIF-1α contributes to in neuroprotection, erythropoiesis, and apoptosis modulation. The purpose of this study was to examine the role played by HIF-1α in regulating neurological injury evoked by intracerebral hemorrhage (ICH) through its downstream product, namely vascular endothelial growth factor (VEGF). In particular, we examined the effects of diabetic hyperglycemia on HIF-1α response in the processing of ICH. METHODS: ELISA was used to measure HIF-1α and VEGF; and Western Blot analysis to examine the protein expression of VEGFR-2 and Caspase-3. Neurological Severity Score and brain water content were used to indicate neurological function and brain edema. RESULTS: HIF-1α and VEGF were significantly increased in the brain after induction of ICH in non-diabetic control rats and diabetic rats; however, the amplified levels of HIF-1α and VEGF were attenuated in diabetic rats (P<0.05 vs. non-diabetic rats) as compared with non-diabetic rats. Also, the protein expression of VEGF receptor subtype 2 was significantly less in the brain of diabetic rats (P<0.05 vs. non-diabetic rats). Further, cerebral infusion of HIF-1 activator stabilized VEGF levels, attenuated Caspase-3 and improved neurological deficits induced by ICH and the effects are smaller in diabetic animals. CONCLUSION: HIF-1α activated by ICH likely plays a beneficial role via VEGF mechanisms and response of HIF-1α is largely impaired in diabetes. This has pharmacological implications to target specific HIF-1α and VEGF pathway for neuronal dysfunction and vulnerability related to ICH.


Asunto(s)
Edema Encefálico/metabolismo , Hemorragia Cerebral/metabolismo , Diabetes Mellitus Experimental/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Encéfalo/metabolismo , Caspasa 3/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Neurochem Res ; 40(6): 1095-101, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25894679

RESUMEN

Capillarisin, one of the major bioactive compounds derived from Artemisia capillaries Thunb, has been reported to have extensive pharmacological properties, such as ant-inflammatory and anti-nociceptive activities. However, the molecular mechanisms responsible for the anti-inflammatory activity of capillarisin have not been elucidated in microglia. In the present study, we investigated the anti-inflammatory effects and molecular mechanisms of capillarisin on LPS-stimulated BV2 microglial cells. The effects of capillarisin on inflammatory mediators TNF-α, IL-6, IL-1ß, NO and PGE2 were detected. The effects of capillarisin on NF-κB and MAPK activation were detected by western blotting. The results showed that capillarisin suppressed LPS-induced TNF-α, IL-6, IL-1ß, NO and PGE2 production in a dose-dependent manner. Capillarisin also inhibited LPS-induced TLR4 expression, NF-κB and MAPKs activation in BV2 microglia. In conclusion, capillarisin inhibited LPS-induced inflammation by blocking TLR4-mediated NF-κB and MAPKs activation in BV2 microglia.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Cromonas/farmacología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/metabolismo , FN-kappa B/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Mediadores de Inflamación/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Óxido Nítrico/metabolismo
17.
Proc Natl Acad Sci U S A ; 109(32): 13112-7, 2012 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-22826221

RESUMEN

Parkinson disease (PD) is a progressive neurodegenerative disease whose progression may be slowed, but at present there is no pharmacological intervention that would stop or reverse the disease. Liver X receptor ß (LXRß) is a member of the nuclear receptor super gene family expressed in the central nervous system, where it is important for cortical layering during development and survival of dopaminergic neurons throughout life. In the present study we have used the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD to investigate the possible use of LXRß as a target for prevention or treatment of PD. The dopaminergic neurons of the substantia nigra of LXRß(-/-) mice were much more severely affected by MPTP than were those of their WT littermates. In addition, the number of activated microglia and GFAP-positive astrocytes was higher in the substantia nigra of LXRß(-/-) mice than in WT littermates. Administration of the LXR agonist GW3965 to MPTP-treated WT mice protected against loss of dopaminergic neurons and of dopaminergic fibers projecting to the striatum, and resulted in fewer activated microglia and astroglia. Surprisingly, LXRß was not expressed in the neurons of the substantia nigra but in the microglia and astroglia. We conclude that LXR agonists may have beneficial effects in treatment of PD by modulating the cytotoxic functions of microglia.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Trastornos Parkinsonianos/metabolismo , Sustancia Negra/citología , Análisis de Varianza , Animales , Astrocitos/metabolismo , Benzoatos/farmacología , Bencilaminas/farmacología , Proteína Ácida Fibrilar de la Glía , Inmunohistoquímica , Receptores X del Hígado , Masculino , Ratones , Ratones Noqueados , Microglía/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Nucleares Huérfanos/antagonistas & inhibidores , Receptores Nucleares Huérfanos/genética , Trastornos Parkinsonianos/prevención & control , Trastornos Parkinsonianos/terapia
18.
Proc Natl Acad Sci U S A ; 109(19): 7493-8, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22529354

RESUMEN

Anxiety disorders are the most prevalent mental disorders in adolescents in the United States. Female adolescents are more likely than males to be affected with anxiety disorders, but less likely to have behavioral and substance abuse disorders. The prefrontal cortex (PFC), amygdala, and dorsal raphe are known to be involved in anxiety disorders. Inhibitory input from the PFC to the amygdala controls fear and anxiety typically originating in the amygdala, and disruption of the inhibitory input from the PFC leads to anxiety, fear, and personality changes. Recent studies have implicated liver X receptor ß (LXRß) in key neurodevelopmental processes and neurodegenerative diseases. In the present study, we used elevated plus-maze, startle and prepulse inhibition, open field, and novel object recognition tests to evaluate behavior in female LXRß KO (LXRß(-/-)) mice. We found that the female LXRß(-/-) mice were anxious with impaired behavioral responses but normal locomotion and memory. Immunohistochemistry analysis revealed decreased expression of the enzyme responsible for GABA synthesis, glutamic acid decarboxylase (65+67), in the ventromedial PFC. Expression of tryptophan hydroxylase 2 in the dorsal raphe was normal. We conclude that the anxiogenic phenotype in female LXRß(-/-) mice is caused by reduced GABAergic input from the ventromedial PFC to the amygdala.


Asunto(s)
Ansiedad/psicología , Glutamato Descarboxilasa/metabolismo , Receptores Nucleares Huérfanos/deficiencia , Corteza Prefrontal/enzimología , Adolescente , Animales , Ansiedad/fisiopatología , Miedo/fisiología , Miedo/psicología , Femenino , Humanos , Inmunohistoquímica , Receptores X del Hígado , Masculino , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/fisiología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Factores Sexuales , Ácido gamma-Aminobutírico/biosíntesis
19.
Proc Natl Acad Sci U S A ; 109(5): 1708-12, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307635

RESUMEN

An estrogen receptor (ER) ß ligand (LY3201) with a preference for ERß over ERα was administered in s.c. pellets releasing 0.04 mg/d. The brains of these mice were examined 3 d after treatment had begun. Although estradiol-17ß is known to increase spine density and glutaminergic signaling, as measured by Golgi staining, a clear reduction in spines was evident on the dendritic branches in LY3201-treated mice but no morphological alteration and no difference in the number of dendritic spines on dendritic stems were observed. In the LY3201-treatment group, there was higher expression of glutamic acid decarboxylase (GAD) in layer V of cortex and in the CA1 of hippocampus, more GAD(+) terminals surrounding the pyramidal neurons and less glutamate receptor (NMDAR) on the neurons in layer V. There were no alterations in expression of Iba1 or in Olig2 or CNPase. However, GFAP(+) astrocytes were increased in the LY3201-treatment group. There were also more projections characteristic of activated astrocytes and increased expression of glutamine synthetase (GS). No expression of ERß was detectable in the nuclei of astrocytes. Clearly, LY3201 caused a shift in the balance between excitatory and inhibitory neurotransmission in favor of inhibition. This shift was due in part to increased synthesis of GABA and increased removal of glutamate from the synaptic cleft by astrocytes. The data reveal that treatment with a selective ERß agonist results in changes opposite to those reported in estradiol-17ß-treated mice and suggests that ERα and ERß play opposing roles in the brain.


Asunto(s)
Benzopiranos/farmacología , Encéfalo/efectos de los fármacos , Espinas Dendríticas/efectos de los fármacos , Receptor beta de Estrógeno/efectos de los fármacos , Transducción de Señal , Ácido gamma-Aminobutírico/metabolismo , Animales , Encéfalo/metabolismo , Espinas Dendríticas/metabolismo , Receptor beta de Estrógeno/metabolismo , Ligandos , Ratones , Receptores de N-Metil-D-Aspartato/metabolismo
20.
Int J Biol Sci ; 19(9): 2848-2859, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37324952

RESUMEN

Liver X receptors (LXRαß) play essential roles in the maintenance of the normal functions of macrophages, in modulation of immune system responses and cholesterol homeostasis. We have reported that LXRαß-/- mice develop squamous cell lung cancer. We now report that those LXRαß-/- mice, which live to 18-months of age, spontaneously develop a second type of lung cancer resembling a rare subtype of NSCLC (TTF-1 and P63-positive). The lesions are characterized as follows: a high proliferation rate; a marked accumulation of abnormal macrophages; an increase in the number of regulatory T cells; a remarkably low level of CD8+ cytotoxic T lymphocytes; enhanced TGFß signaling; an increased expression of matrix metalloproteinases accompanied by degradation of lung collagen; and a loss of estrogen receptor ß (ERß). Because NSCLC is associated with cigarette smoking, we investigated the possible links between loss of LXRαß and CS. A Kaplan-Meier Plotter database revealed reduced expression of LXRαß and ERß was correlated with low overall survival (OS). Thus, reduction of LXRαß expression by cigarette smoking may be one mechanism through which CS causes lung cancer. The possibility that maintenance of LXRαß and ERß signaling could be used in the treatment of NSCLC needs further investigation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Ratones , Animales , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Receptores X del Hígado/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA