Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(6): 284, 2022 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-35526196

RESUMEN

BACKGROUND AND AIMS: Recent evidences highlight a role of the mitochondria calcium homeostasis in the development of colorectal cancer (CRC). To overcome treatment resistance, we aimed to evaluate the role of the mitochondrial sodium-calcium-lithium exchanger (NCLX) and its targeting in CRC. We also identified curcumin as a new inhibitor of NCLX. METHODS: We examined whether curcumin and pharmacological compounds induced the inhibition of NCLX-mediated mitochondrial calcium (mtCa2+) extrusion, the role of redox metabolism in this process. We evaluated their anti-tumorigenic activity in vitro and in a xenograft mouse model. We analyzed NCLX expression and associations with survival in The Cancer Genome Atlas (TCGA) dataset and in tissue microarrays from 381 patients with microsatellite instability (MSI)-driven CRC. RESULTS: In vitro, curcumin exerted strong anti-tumoral activity through its action on NCLX with mtCa2+ and reactive oxygen species overload associated with a mitochondrial membrane depolarization, leading to reduced ATP production and apoptosis. NCLX inhibition with pharmacological and molecular approaches reproduced the effects of curcumin. NCLX inhibitors decreased CRC tumor growth in vivo. Both transcriptomic analysis of TCGA dataset and immunohistochemical analysis of tissue microarrays demonstrated that higher NCLX expression was associated with MSI status, and for the first time, NCLX expression was significantly associated with recurrence-free survival. CONCLUSIONS: Our findings highlight a novel anti-tumoral mechanism of curcumin through its action on NCLX and mitochondria calcium overload that could benefit for therapeutic schedule of patients with MSI CRC.


Asunto(s)
Neoplasias Colorrectales , Curcumina , Inestabilidad de Microsatélites , Intercambiador de Sodio-Calcio , Animales , Calcio/metabolismo , Señalización del Calcio , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Curcumina/farmacología , Humanos , Ratones , Repeticiones de Microsatélite , Proteínas Mitocondriales/metabolismo , Intercambiador de Sodio-Calcio/antagonistas & inhibidores
2.
J Nutr ; 151(5): 1119-1127, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33831951

RESUMEN

BACKGROUND: Several studies have recently highlighted important roles for adipose tissue in cancer. However, few have examined adipose tissue cholesterol, and no study has been performed in breast adipose tissue associated with breast tumors. OBJECTIVES: The present work was designed to determine if breast adipose tissue cholesterol from the tumor-surrounding area is associated with breast cancer aggressiveness. METHODS: Between 2009 and 2011, 215 breast adipose tissue samples were collected at the Tours University Hospital (France) during surgery of women (aged 28-89 y) with invasive breast cancer. Associations of free cholesterol (FC), esterified cholesterol (EC), and total cholesterol (TC) amounts with clinical variables (age, BMI, and treated or untreated hypercholesterolemia) and tumor aggressiveness parameters [phenotype, grade, presence of inflammatory breast cancer (IBC), and multifocality] were tested using Student's t test and after ANOVA. RESULTS: The predominant form of cholesterol in adipose tissue was FC, and 50% of patients had no detectable EC. The adipose tissue FC content (µg/mg total lipid) was 18% greater in patients >70 y old than in those 40-49 y old (P < 0.05) and the TC content tended to be 12% greater in untreated hypercholesterolemic patients than in normocholesterolemic patients (P = 0.06). Breast adipose cholesterol concentrations were increased in tissues obtained from patients with human-epidermal-growth-factor-receptor-2 (HER2) phenotype (+13% FC; P < 0.05 compared with luminal A), IBC (+15% FC; P = 0.06 compared with noninflammatory tumors), as well as with multifocal triple-negative tumors (+34% FC, P < 0.05; +30% TC, P < 0.05, compared with unifocal triple-negative tumors). Among patients with triple-negative tumors, hypercholesterolemia was significantly more common (P < 0.05) in patients with multifocal tumors (64%) than in patients with unifocal tumors (25%). CONCLUSIONS: This study is the first of this magnitude that analyzes cholesterol concentrations in adipose tissue from female breast cancer patients. An increase in breast adipose tissue cholesterol content may contribute to breast cancer aggressiveness (HER2 phenotype, multifocality of triple-negative tumors, and IBC).


Asunto(s)
Tejido Adiposo/metabolismo , Neoplasias de la Mama/patología , Colesterol/metabolismo , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Invasividad Neoplásica/patología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/epidemiología , Femenino , Francia/epidemiología , Humanos , Persona de Mediana Edad
3.
Breast Cancer Res ; 22(1): 38, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32321558

RESUMEN

BACKGROUND: The highest incidence of breast cancer is in the Western world. Several aspects of the Western lifestyle are known risk factors for breast cancer. In particular, previous studies have shown that cholesterol levels can play an important role in the regulation of tumor progression. METHODS: In the present study, we modulated cholesterol metabolism in the human breast cancer cell lines MCF-7 and MDA-MB-231 using a genetic approach. Apolipoprotein A-I (apoA-I) and apolipoprotein E (apoE) were expressed in these cell lines to modulate cholesterol metabolism. The effects of these apolipoproteins on cancer cell properties were examined. RESULTS: Our results show that both apolipoproteins can regulate cholesterol metabolism and can control the epithelial-to-mesenchymal transition process. However, these effects were different depending on the cell type. We show that expressing apoA-I or apoE stimulates proliferation, migration, and tumor growth of MCF-7 cells. However, apoA-I or apoE reduces proliferation and migration of MDA-MB-231 cells. CONCLUSIONS: These data suggest that modulating sterol metabolism may be most effective at limiting tumor progression in models of triple-negative cancers.


Asunto(s)
Apolipoproteína A-I/metabolismo , Apolipoproteínas E/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Colesterol/metabolismo , Metabolismo de los Lípidos , Animales , Neoplasias de la Mama/clasificación , Línea Celular Tumoral , Movimiento Celular , Bases de Datos Genéticas , Transición Epitelial-Mesenquimal , Femenino , Humanos , Ratones , Ratones Desnudos , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Int J Mol Sci ; 21(14)2020 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-32674321

RESUMEN

n-3 long chain Polyunsaturated Fatty Acids (n-3 LCPUFA) have been shown to improve the efficacy of conventional chemotherapies used for breast cancer treatment. In addition to their reported ability to increase the chemosensitivity of cancer cells, we hypothesized that n-3 LCPUFA could induce a remodeling of the vascular network in mammary tumors. A contrast-enhanced ultrasound method was used to monitor the vascular architecture during docetaxel treatment of mammary tumors in rats fed either a control or an n-3 LCPUFA-enriched diet (docosahexaenoic acid (DHA)/eicosapentaenoic acid (EPA)). The vascular network was remodeled in favor of smaller vessels (microvascularization), which represented 54% of the vasculature in n-3 LCPUFA tumors but only 26% in control tumors after 2 weeks of chemotherapy. Importantly, vascularization changes occurred both before and during docetaxel treatment. The density of smaller vessels quantified before chemotherapy was correlated with improved tumor size reduction by docetaxel treatment. Furthermore, transcript levels of the angiogenesis-specific genes epiregulin and amphiregulin were reduced by ~4.5- and twofold in tumors obtained from rats fed an n-3 LCPUFA-enriched diet compared to those of rats fed a control diet, respectively. Their expression levels were negatively correlated with tumor regression after chemotherapy. Taken together, this preclinical data strengthen the potential usefulness of n-3 LCPUFA as a complementary clinical strategy to improve drug efficiency via remodeling of the tumor vasculature.


Asunto(s)
Docetaxel/farmacología , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Ácidos Grasos/farmacología , Neoplasias Mamarias Animales/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Animales , Dieta , Ácidos Grasos Omega-3/farmacología , Femenino , Neoplasias Mamarias Animales/patología , Neovascularización Patológica/metabolismo , Ratas , Ratas Sprague-Dawley
5.
J Biol Chem ; 288(5): 3477-88, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23258544

RESUMEN

Insulin resistance, hyperlipidemia, and cardiovascular complications are common dysregulations of metabolic syndrome. Transplant patients treated with immunosuppressant drugs such as cyclosporine A (CsA), an inhibitor of calcineurin phosphatase, frequently develop similar metabolic complications. Although calcineurin is known to mediate insulin sensitivity by regulating ß-cell growth and adipokine gene transcription, its role in lipid homeostasis is poorly understood. Here, we examined lipid homeostasis in mice lacking calcineurin Aß (CnAß(-/-)). We show that mice lacking calcineurin Aß are hyperlipidemic and develop age-dependent insulin resistance. Hyperlipidemia found in CnAß(-/-) mice is, in part, due to increased lipolysis in adipose tissues, a process mediated by ß-adrenergic G-protein-coupled receptor signaling pathways. CnAß(-/-) mice also exhibit additional pathophysiological phenotypes caused by the potentiated GPCR signaling pathways. A cell autonomous mechanism with sustained cAMP/PKA activation is found in CnAß(-/-) mice or upon CsA treatment to inhibit calcineurin. Increased PKA activation and cAMP accumulation in CnAß(-/-) mice, however, are sensitive to phosphodiesterase inhibitor. Indeed, we show that calcineurin regulates degradation of phosphodiesterase 3B, in addition to phosphodiesterase 4D. These results establish a role for calcineurin in lipid homeostasis. These data also indicate that potentiated cAMP signaling pathway may provide an alternative molecular pathogenesis for the metabolic complications elicited by CsA in transplant patients.


Asunto(s)
Calcineurina/deficiencia , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Hiperlipidemias/enzimología , Transducción de Señal , Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Secuencia de Aminoácidos , Animales , Células COS , Calcineurina/metabolismo , Chlorocebus aethiops , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/química , Ciclosporina/farmacología , Embrión de Mamíferos/citología , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Hiperlipidemias/patología , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Inhibidores de Fosfodiesterasa/farmacología , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/efectos de los fármacos , Triglicéridos/biosíntesis
6.
Cell Tissue Res ; 356(1): 147-57, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24390341

RESUMEN

Clinical studies have established the important impact of atherosclerotic disease in Western societies. This disease is characterized by the accumulation of lipids and the migration of various cell types in the sub-endothelial space of blood vessels. As demonstrated by many studies, endothelial cells play an essential role in the development of this disease. The endothelium acts as a gatekeeper of blood vessel integrity and cardiovascular health status. For instance, the transfer of lipids via the transport of lipoproteins in the arterial intima is believed to be mediated by endothelial cells through a process termed transcytosis. In addition, lipoproteins that accumulate in the sub-endothelial space may also be modified, in a process that can direct the activation of endothelial cells. These steps are essential for the initiation of an atherosclerotic plaque and may be mediated, at least in part, by caveolae and their associated protein caveolin-1. In the present study, we evaluate the role of caveolin-1/caveolae in the regulation of these two steps in endothelial cells. Our data clearly demonstrate that caveolin-1 is involved in the regulation of lipoprotein transcytosis across endothelial cells and in the regulation of vascular inflammation.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/patología , Caveolina 1/metabolismo , Células Endoteliales/metabolismo , Albúminas/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Caveolas/metabolismo , Regulación hacia Abajo , Endocitosis , Células Endoteliales/patología , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Inflamación/patología , Lipoproteínas LDL/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
7.
Cell Tissue Res ; 358(3): 821-31, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25322709

RESUMEN

Atherosclerosis is a complex disease initiated by the vascular accumulation of lipoproteins in the sub-endothelial space, followed by the infiltration of monocytes into the arterial intima. Caveolin-1 (Cav-1) plays an essential role in the regulation of cellular cholesterol metabolism and of various signaling pathways. In order to study specifically the role of macrophage Cav-1 in atherosclerosis, we used Cav-1 (-/-) Apoe (-/-) mice and transplanted them with bone marrow (BM) cells obtained from Cav-1 (+/+) Apoe (-/-) or Cav-1 (-/-) Apoe (-/-) mice and vice versa. We found that Cav-1 (+/+) mice harboring Cav-1 (-/-) BM-derived macrophages developed significantly larger lesions than Cav-1 (+/+) mice harboring Cav-1 (+/+) BM-derived macrophages. Cav-1 (-/-) macrophages were more susceptible to apoptosis and more prone to induce inflammation. The present study provides clear evidence that the absence of Cav-1 in macrophage is pro-atherogenic, whereas its absence in endothelial cells protects against atherosclerotic lesion formation. These findings demonstrate the cell-specific role of Cav-1 during the development of this disease.


Asunto(s)
Aterosclerosis/patología , Caveolina 1/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Animales , Apoptosis/efectos de los fármacos , Aterosclerosis/sangre , Trasplante de Médula Ósea , Caveolina 1/deficiencia , Citocinas/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Lipoproteínas/sangre , Macrófagos Peritoneales/efectos de los fármacos , Ratones Endogámicos C57BL , Regulación hacia Arriba/efectos de los fármacos
8.
Breast Cancer Res ; 15(5): R87, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24060386

RESUMEN

INTRODUCTION: Previous studies have identified cholesterol as an important regulator of breast cancer development. High-density lipoprotein (HDL) and its cellular receptor, the scavenger receptor class B type I (SR-BI) have both been implicated in the regulation of cellular cholesterol homeostasis, but their functions in cancer remain to be established. METHODS: In the present study, we have examined the role of HDL and SR-BI in the regulation of cellular signaling pathways in breast cancer cell lines and in the development of tumor in a mouse xenograft model. RESULTS: Our data show that HDL is capable of stimulating migration and can activate signal transduction pathways in the two human breast cancer cell lines, MDA-MB-231 and MCF7. Furthermore, we also show that knockdown of the HDL receptor, SR-BI, attenuates HDL-induced activation of the phosphatidylinositol 3-kinase (PI3K)/protein Kinase B (Akt) pathway in both cell lines. Additional investigations show that inhibition of the PI3K pathway, but not that of the mitogen-activated protein kinase (MAPK) pathway, could lead to a reduction in cellular proliferation in the absence of SR-BI. Importantly, whereas the knockdown of SR-BI led to decreased proliferation and migration in vitro, it also led to a significant reduction in tumor growth in vivo. Most important, we also show that pharmacological inhibition of SR-BI can attenuate signaling and lead to decreased cellular proliferation in vitro. Taken together, our data indicate that both cholesteryl ester entry via HDL-SR-BI and Akt signaling play an essential role in the regulation of cellular proliferation and migration, and, eventually, tumor growth. CONCLUSIONS: These results identify SR-BI as a potential target for the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Antígenos CD36/metabolismo , Transformación Celular Neoplásica , Colesterol/metabolismo , Transducción de Señal , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Antígenos CD36/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Colesterol/farmacología , HDL-Colesterol/metabolismo , HDL-Colesterol/farmacología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Células MCF-7 , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Lipoproteína/genética , Receptores de Lipoproteína/metabolismo , Carga Tumoral/genética
9.
J Steroid Biochem Mol Biol ; 232: 106346, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37321513

RESUMEN

Cholesterol plays important roles in many physiological processes, including cell membrane structure and function, hormone synthesis, and the regulation of cellular homeostasis. The role of cholesterol in breast cancer is complex, and some studies have suggested that elevated cholesterol levels may be associated with an increased risk of developing breast cancer, while others have found no significant association. On the other hand, other studies have shown that, for total cholesterol and plasma HDL-associated cholesterol levels, there was inverse association with breast cancer risk. One possible mechanism by which cholesterol may contribute to breast cancer risk is as a key precursor of estrogen. Other potential mechanisms by which cholesterol may contribute to breast cancer risk include its role in inflammation and oxidative stress, which have been linked to cancer progression. Cholesterol has also been shown to play a role in signaling pathways regulating the growth and proliferation of cancer cells. In addition, recent studies have shown that cholesterol metabolism can generate tumor promoters such as cholesteryl esters, oncosterone, 27-hydroxycholesterol but also tumor suppressor metabolites such as dendrogenin A. This review summarizes some of the most important clinical studies that have evaluated the role of cholesterol or its derivatives in breast cancer. It also addresses the role of cholesterol and its derivatives at the cellular level.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/metabolismo , Incidencia , Colesterol/metabolismo , Ésteres del Colesterol/metabolismo , Factores de Riesgo
10.
Am J Pathol ; 178(1): 402-12, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224077

RESUMEN

Diet and obesity are important risk factors for cancer development. Many studies have suggested an important role for several dietary nutrients in the progression and development of breast cancer. However, few studies have specifically addressed the role of components of a Western diet as important factors involved in breast cancer initiation and progression. The present study examined the role of cholesterol in the regulation of tumor progression in a mouse model of mammary tumor formation. The results suggest that cholesterol accelerates and enhances tumor formation. In addition, tumors were more aggressive, and tumor angiogenesis was enhanced. Metabolism of cholesterol was also examined in this mouse model. It was observed that plasma cholesterol levels were reduced during tumor development but not prior to its initiation. These data provide new evidence for an increased utilization of cholesterol by tumors and for its role in tumor formation. Taken together, these results imply that an increase in plasma cholesterol levels accelerates the development of tumors and exacerbates their aggressiveness.


Asunto(s)
Neoplasias de la Mama/etiología , Transformación Celular Neoplásica , Colesterol/efectos adversos , Dieta/efectos adversos , Neoplasias Mamarias Experimentales/etiología , Animales , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Colesterol/administración & dosificación , Colesterol/sangre , Femenino , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/sangre , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Biosíntesis de Proteínas
11.
Adv Exp Med Biol ; 729: 127-44, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22411318

RESUMEN

Atherosclerosis is a disease of the blood vessel characterized by the development of an arterial occlusion containing lipid and cellular deposits. Caveolae are 50-100 nm cell surface plasma membrane invaginations that are believed to play an important role in the regulation of cellular signaling and transport of molecules among others. These organelles are enriched in sphingolipids and cholesterol and are characterized by the presence of the protein caveolin-1. Caveolin-1 and caveolae are present in most of the cells involved in the development of atherosclerosis. The current literature suggests a rather complex role for caveolin-1 in this disease, with evidence of either pro- or anti-atherogenic functions depending on the cell type examined. In the present chapter, the various roles of caveolae and caveolin-1 in the development of atherosclerosis are examined.


Asunto(s)
Aterosclerosis , Caveolas , Caveolina 1 , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Caveolas/metabolismo , Caveolina 1/química , Caveolina 1/deficiencia , Caveolina 1/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología
13.
Am J Pathol ; 177(6): 3180-91, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21088217

RESUMEN

Epidemiological studies have provided evidence suggesting an important role for diet and obesity in the development of cancer. Specifically, lipid nutrients of the diet have been identified as important regulators of tumor development and progression. In the present study, we have examined the role of dietary fat and cholesterol in the initiation and progression of prostate cancer using the well-characterized TRAMP mouse model. Consumption of a Western-type diet--that is, enriched in both fat and cholesterol--accelerated prostate tumor incidence and tumor burden compared to mice fed a control chow diet. Furthermore, we also show that this diet increased the extent and the histological grade of prostate tumors. These findings were confirmed by the presence of increased levels of protein markers of advanced tumors in prostates obtained from animals fed a Western-type diet compared to those obtained from control animals. Increased lung metastases in animals fed a Western-type diet were also observed. In addition, we found that with a Western diet, animals bearing tumors presented with reduced plasma cholesterol levels compared with animals fed a control diet. Finally, we show that tumors obtained from animals fed a Western-type diet displayed increased expression of the high-density lipoprotein receptor SR-BI and increased angiogenesis. Taken together, our data suggest that dietary fat and cholesterol play an important role in the development of prostate cancer.


Asunto(s)
Adenocarcinoma/patología , Dieta/efectos adversos , Neoplasias de la Próstata/patología , Adenocarcinoma/sangre , Adenocarcinoma/etiología , Adenocarcinoma/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/metabolismo , Carga Tumoral , Mundo Occidental
14.
Mol Genet Metab ; 104(3): 346-55, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21724437

RESUMEN

The Smith-Lemli-Opitz syndrome (SLOS) is an inherited disorder of cholesterol synthesis caused by mutations in DHCR7 which encodes the final enzyme in the cholesterol synthesis pathway. The immediate precursor to cholesterol synthesis, 7-dehydrocholesterol (7-DHC) accumulates in the plasma and cells of SLOS patients which has led to the idea that the accumulation of abnormal sterols and/or reduction in cholesterol underlies the phenotypic abnormalities of SLOS. We tested the hypothesis that 7-DHC accumulates in membrane caveolae where it disturbs caveolar bilayer structure-function. Membrane caveolae from skin fibroblasts obtained from SLOS patients were isolated and found to accumulate 7-DHC. In caveolar-like model membranes containing 7-DHC, subtle, but complex alterations in intermolecular packing, lipid order and membrane width were observed. In addition, the BK(Ca) K(+) channel, which co-migrates with caveolin-1 in a membrane fraction enriched with cholesterol, was impaired in SLOS cells as reflected by reduced single channel conductance and a 50 mV rightward shift in the channel activation voltage. In addition, a marked decrease in BK(Ca) protein but not mRNA expression levels was seen suggesting post-translational alterations. Accompanying these changes was a reduction in caveolin-1 protein and mRNA levels, but membrane caveolar structure was not altered. These results are consistent with the hypothesis that 7-DHC accumulation in the caveolar membrane results in defective caveolar signaling. However, additional cellular alterations beyond mere changes associated with abnormal sterols in the membrane likely contribute to the pathogenesis of SLOS.


Asunto(s)
Caveolas/metabolismo , Deshidrocolesteroles/metabolismo , Fibroblastos/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Síndrome de Smith-Lemli-Opitz/metabolismo , Caveolina 1/metabolismo , Células Cultivadas , Deshidrocolesteroles/química , Genotipo , Humanos , Immunoblotting , Membranas Artificiales , Microscopía Electrónica , Estructura Molecular , Piel/citología , Esteroles/metabolismo , Difracción de Rayos X
15.
Am J Physiol Cell Physiol ; 298(3): C693-701, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20007452

RESUMEN

The goal of this study was to determine whether caveolin-2 (Cav-2) is capable of controlling endothelial cell (EC) proliferation in vitro. To realize this goal, we have directly compared proliferation rates and cell cycle-associated signaling proteins between lung ECs isolated from wild-type (WT) and Cav-2 knockout (KO) mice. Using three independent proliferation assays, we have determined that Cav-2 KO ECs proliferate by ca. 2-fold faster than their WT counterparts. Cell cycle analysis by flow cytometry of propidium iodide-stained cells showed a relatively higher percentage of Cav-2 KO ECs in S and G(2)/M and lower percentage in G(o)/G(1) phases of cell cycle relative to their WT counterparts. Furthermore, an over 2-fold increase in the percentage of S phase-associated Cav-2 KO relative to WT ECs was independently determined with bromodeoxyuridine incorporation assay. Mechanistically, the increase in proliferation/cell cycle progression of Cav-2 KO ECs correlated well with elevated expression levels of predominantly S phase- and G(2)/M phase-associated cyclin A and B1, respectively. Further mechanistic analysis of molecular events controlling cell cycle progression revealed increased level of hyperphosphorylated (inactive) form of G(1) to S phase transition inhibitor, the retinoblastoma protein in hyperproliferating Cav-2 KO ECs. Conversely, the expression level of the two cyclin-dependent kinase inhibitors p16(INK4) and p27(Kip1) was reduced in Cav-2 KO ECs. Finally, increased phosphorylation (activation) of proproliferative extracellular signal-regulated kinase 1/2 was observed in hyperproliferating Cav-2 KO ECs. Overall, our data suggest that Cav-2 negatively regulates lung EC proliferation and cell cycle progression.


Asunto(s)
Caveolina 2/deficiencia , Ciclo Celular , Proliferación Celular , Células Endoteliales/metabolismo , Animales , Caveolina 2/genética , Células Cultivadas , Ciclina A/metabolismo , Ciclina B1/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Citometría de Flujo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pulmón/irrigación sanguínea , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Factores de Tiempo
16.
Am J Pathol ; 174(3): 746-61, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19234134

RESUMEN

Recently, we reported that human breast cancer-associated fibroblasts show functional inactivation of the retinoblastoma (RB) tumor suppressor and down-regulation of caveolin-1 (Cav-1) protein expression. However, it remains unknown whether loss of Cav-1 is sufficient to confer functional RB inactivation in mammary fibroblasts. To establish a direct cause-and-effect relationship, mammary stromal fibroblasts (MSFs) were prepared from Cav-1(-/-) null mice and subjected to phenotypic analysis. Here, we provide evidence that Cav-1(-/-) MSFs share many characteristics with human cancer-associated fibroblasts. The Cav-1(-/-) MSF transcriptome significantly overlaps with human cancer-associated fibroblasts; both show a nearly identical profile of RB/E2F-regulated genes that are up-regulated, which is consistent with RB inactivation. This Cav-1(-/-) MSF gene signature is predictive of poor clinical outcome in breast cancer patients treated with tamoxifen. Consistent with these findings, Cav-1(-/-) MSFs show RB hyperphosphorylation and the up-regulation of estrogen receptor co-activator genes. We also evaluated the paracrine effects of "conditioned media" prepared from Cav-1(-/-) MSFs on wild-type mammary epithelia. Our results indicate that Cav-1(-/-) MSF "conditioned media" is sufficient to induce an epithelial-mesenchymal transition, indicative of an invasive phenotype. Proteomic analysis of this "conditioned media" reveals increased levels of proliferative/angiogenic growth factors. Consistent with these findings, Cav-1(-/-) MSFs are able to undergo endothelial-like transdifferentiation. Thus, these results have important implications for understanding the role of cancer-associated fibroblasts and RB inactivation in promoting tumor angiogenesis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Caveolina 1/deficiencia , Caveolina 1/genética , Fibroblastos/patología , Células del Estroma/patología , Western Blotting , Mama/citología , Mama/fisiología , Neoplasias de la Mama/mortalidad , Técnicas de Cultivo de Célula , División Celular , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Células Epiteliales/citología , Células Epiteliales/patología , Células Epiteliales/fisiología , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Neoplásico/genética , Células del Estroma/citología , Células del Estroma/fisiología , Análisis de Supervivencia
17.
Am J Pathol ; 174(2): 613-29, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19164602

RESUMEN

Here, we show that functional loss of a single gene is sufficient to confer constitutive milk protein production and protection against mammary tumor formation. Caveolin-3 (Cav-3), a muscle-specific caveolin-related gene, is highly expressed in muscle cells. We demonstrate that Cav-3 is also expressed in myoepithelial cells within the mammary gland. To determine whether genetic ablation of Cav-3 expression affects adult mammary gland development, we studied the phenotype(s) of Cav-3(-/-)-null mice. Interestingly, Cav-3(-/-) virgin mammary glands developed lobulo-alveolar hyperplasia, akin to the changes normally observed during pregnancy and lactation. Genome-wide expression profiling revealed up-regulation of gene transcripts associated with pregnancy/lactation, mammary stem cells, and human breast cancers, consistent with a constitutive lactogenic phenotype. Expression levels of three key transcriptional regulators of lactation, namely Elf5, Stat5a, and c-Myc, were also significantly elevated. Experiments with pregnant mice directly showed that Cav-3(-/-) mice underwent precocious lactation. Finally, using orthotopic tumor cell implantation, we demonstrated that virgin Cav-3(-/-) mice were dramatically protected against mammary tumor formation. Thus, Cav-3(-/-) mice are a novel preclinical model to study the protective effects of a lactogenic microenvironment on mammary tumor onset and progression. Our current studies have broad implications for using the lactogenic microenvironment as a paradigm to discover new therapies for the prevention and/or treatment of human breast cancers.


Asunto(s)
Caveolina 3/genética , Caveolina 3/metabolismo , Expresión Génica , Lactancia/fisiología , Neoplasias Mamarias Experimentales/genética , Animales , Movimiento Celular/fisiología , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Mutantes , Leche Humana/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Reacción en Cadena de la Polimerasa , Embarazo
18.
Am J Pathol ; 174(4): 1172-90, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19342371

RESUMEN

Caveolin-1 (Cav-1) loss-of-function mutations are exclusively associated with estrogen receptor-positive (ER(+)) human breast cancers. To dissect the role of Cav-1 loss-of-function in the pathogenesis of human breast cancers, we used Cav-1(-/-) null mice as a model system. First, we demonstrated that Cav-1(-/-) mammary epithelia overexpress two well-established ER co-activator genes, CAPER and Foxa1, in addition to ER-alpha. Thus, the functional loss of Cav-1 may be sufficient to confer estrogen-hypersensitivity in the mammary gland. To test this hypothesis directly, we subjected Cav-1(-/-) mice to ovariectomy and estrogen supplementation. As predicted, Cav-1(-/-) mammary glands were hyper-responsive to estrogen and developed dysplastic mammary lesions with adjacent stromal angiogenesis that resemble human ductal carcinoma in situ. Based on an extensive biomarker analysis, these Cav-1(-/-) mammary lesions contain cells that are hyperproliferative and stain positively with nucleolar (B23/nucleophosmin) and stem/progenitor cell markers (SPRR1A and beta-catenin). Genome-wide transcriptional profiling identified many estrogen-related genes that were over-expressed in Cav-1(-/-) mammary glands, including CAPER--an ER co-activator gene and putative stem/progenitor cell marker. Analysis of human breast cancer samples revealed that CAPER is overexpressed and undergoes a cytoplasmic-to-nuclear shift during the transition from pre-malignancy to ductal carcinoma in situ. Thus, Cav-1(-/-) null mice are a new preclinical model for studying the molecular paradigm of estrogen hypersensitivity and the development of estrogen-dependent ductal carcinoma in situ lesions.


Asunto(s)
Carcinoma Intraductal no Infiltrante/genética , Caveolina 1/genética , Estrógenos/farmacología , Perfilación de la Expresión Génica , Neoplasias Mamarias Experimentales/genética , Animales , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Caveolina 1/deficiencia , Transformación Celular Neoplásica/genética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Inmunohistoquímica , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovariectomía , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Análisis de Matrices Tisulares , Transactivadores/genética , Transactivadores/metabolismo
19.
Am J Pathol ; 174(5): 1650-62, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19395651

RESUMEN

Here we used the Met-1 cell line in an orthotopic transplantation model in FVB/N mice to dissect the role of the Cav-1(P132L) mutation in human breast cancer. Identical experiments were performed in parallel with wild-type Cav-1. Cav-1(P132L) up-regulated the expression of estrogen receptor-alpha as predicted, because only estrogen receptor-alpha-positive patients have been shown to harbor Cav-1(P132L) mutations. In the context of primary tumor formation, Cav-1(P132L) behaved as a loss-of-function mutation, lacking any tumor suppressor activity. In contrast, Cav-1(P132L) caused significant increases in cell migration, invasion, and experimental metastasis, consistent with a gain-of-function mutation. To identify possible molecular mechanism(s) underlying this invasive gain-of-function activity, we performed unbiased gene expression profiling. From this analysis, we show that the Cav-1(P132L) expression signature contains numerous genes that have been previously associated with cell migration, invasion, and metastasis. These include i) secreted growth factors and extracellular matrix proteins (Cyr61, Plf, Pthlh, Serpinb5, Tnc, and Wnt10a), ii) proteases that generate EGF and HGF (Adamts1 and St14), and iii) tyrosine kinase substrates and integrin signaling/adapter proteins (Akap13, Cdcp1, Ddef1, Eps15, Foxf1a, Gab2, Hs2st1, and Itgb4). Several of the P132L-specific genes are also highly expressed in stem/progenitor cells or are associated with myoepithelial cells, suggestive of an epithelial-mesenchymal transition. These results directly support clinical data showing that patients harboring Cav-1 mutations are more likely to undergo recurrence and metastasis.


Asunto(s)
Biomarcadores de Tumor/genética , Caveolina 1/genética , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Mutación/genética , Células Madre Neoplásicas/patología , Animales , Biomarcadores de Tumor/metabolismo , Western Blotting , Caveolina 1/metabolismo , Movimiento Celular , Proliferación Celular , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Neoplasias Mamarias Animales/metabolismo , Ratones , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Transducción de Señal
20.
J Surg Res ; 163(2): e113-22, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20538289

RESUMEN

BACKGROUND: Increased expression of cyclooxygenase (COX-2) contributes to atherosclerosis. Recent studies suggest that COX-2 inhibitors prevent early plaque development but their effects on established lesions are less clear, while the statins promote plaque stability. The purpose of this study is to investigate whether administering a combination of a COX-2 inhibitor with a statin drug alters plaque progression in apo E-/- mice. MATERIALS AND METHODS: Apo E-/- mice were fed a Western diet from 6 to 26 wk of age. At 26 wk, the Western diets supplemented with atorvastatin, celecoxib, or atorvastatin plus celecoxib were given for an additional 12 wk. RESULTS: When the mice were 38 wk of age, the total area occupied by the atherosclerotic lesion was 53% less in the mice fed the combination of atorvastatin + celecoxib P ≤ 0.05) than that of the apo E-/- mice fed the Western diet alone, atorvastatin alone, or celecoxib alone. The decreased extent of atherosclerosis observed in the apo E-/- mice fed the combination of drugs was associated with reduced levels of prostaglandin (PG) E(2,) decreased protein expression of metalloproteinase (MMP)-9, macrophage chemotactic protein (MCP-1), and COX 2, and decreased staining for MMP-9, F4-80 (a marker for macrophages), and vascular cell adhesion molecule (VCAM). CONCLUSION: This study indicates that using statins with a COX-2 inhibitor reduced the extent of atherosclerosis and inflammatory/cell adhesion molecule levels in the apo E-/- mouse model.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Ácidos Heptanoicos/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Pirazoles/administración & dosificación , Pirroles/administración & dosificación , Sulfonamidas/administración & dosificación , Animales , Apolipoproteínas E/fisiología , Atorvastatina , Celecoxib , Quimiocina CCL2/sangre , Ciclooxigenasa 2/sangre , Dinoprostona/sangre , Progresión de la Enfermedad , Quimioterapia Combinada , Inmunohistoquímica , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Ratones , Ratones Endogámicos C57BL , Molécula 1 de Adhesión Celular Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA