Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Clin Cancer Res ; 29(5): 866-877, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36269797

RESUMEN

PURPOSE: Preclinical data suggest that antiprogestins inhibit the growth of luminal breast carcinomas that express higher levels of progesterone receptor isoform A (PRA) than isoform B (PRB). Thus, we designed a presurgical window of opportunity trial to determine the therapeutic effects of mifepristone in patients with breast cancer, based on their high PRA/PRB isoform ratio (MIPRA; NCT02651844). PATIENTS AND METHODS: Twenty patients with luminal breast carcinomas with PRA/PRB > 1.5 (determined by Western blots), and PR ≥ 50%, naïve from previous treatment, were included for mifepristone treatment (200 mg/day orally; 14 days). Core needle biopsies and surgical samples were formalin fixed for IHC studies, while others were snap-frozen to perform RNA sequencing (RNA-seq), proteomics, and/or Western blot studies. Plasma mifepristone levels were determined using mass spectrometry. The primary endpoint was the comparison of Ki67 expression pretreatment and posttreatment. RESULTS: A 49.62% decrease in Ki67 staining was observed in all surgical specimens compared with baseline (P = 0.0003). Using the prespecified response parameter (30% relative reduction), we identified 14 of 20 responders. Mifepristone induced an increase in tumor-infiltrating lymphocytes; a decrease in hormone receptor and pSer118ER expression; and an increase in calregulin, p21, p15, and activated caspase 3 expression. RNA-seq and proteomic studies identified downregulated pathways related to cell proliferation and upregulated pathways related to immune bioprocesses and extracellular matrix remodeling. CONCLUSIONS: Our results support the use of mifepristone in patients with luminal breast cancer with high PRA/PRB ratios. The combined effects of mifepristone and estrogen receptor modulators warrant clinical evaluation to improve endocrine treatment responsiveness in these patients. See related commentary by Ronchi and Brisken, p. 833.


Asunto(s)
Neoplasias de la Mama , Mifepristona , Humanos , Femenino , Mifepristona/farmacología , Mifepristona/uso terapéutico , Receptores de Progesterona/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteómica , Antígeno Ki-67 , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
2.
Cancer Res ; 81(5): 1375-1387, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33268529

RESUMEN

The role of active antitumor immunity in hormone receptor-positive (HR+) breast cancer has been historically underlooked. The aim of this study was to determine the contribution of the immune system to antiprogestin-induced tumor growth inhibition using a hormone-dependent breast cancer model. BALB/c-GFP+ bone marrow (BM) cells were transplanted into immunodeficient NSG mice to generate an immunocompetent NSG/BM-GFP+ (NSG-R) mouse model. Treatment with the antiprogestin mifepristone (MFP) inhibited growth of 59-2-HI tumors with similar kinetics in both animal models. Interestingly, MFP treatment reshaped the tumor microenvironment, enhancing the production of proinflammatory cytokines and chemokines. Tumors in MFP-treated immunocompetent mice showed increased infiltration of F4/80+ macrophages, natural killer, and CD8 T cells, displaying a central memory phenotype. Mechanistically, MFP induced immunogenic cell death (ICD) in vivo and in vitro, as depicted by the expression and subcellular localization of the alarmins calreticulin and HMGB-1 and the induction of an ICD gene program. Moreover, MFP-treated tumor cells efficiently activated immature dendritic cells, evidenced by enhanced expression of MHC-II and CD86, and induced a memory T-cell response, attenuating tumor onset and growth after re-challenge. Finally, MFP treatment increased the sensitivity of HR+ 59-2-HI tumor to PD-L1 blockade, suggesting that antiprogestins may improve immunotherapy response rates. These results contribute to a better understanding of the mechanisms underlying the antitumor effect of hormonal treatment and the rational design of therapeutic combinations based on endocrine and immunomodulatory agents in HR+ breast cancer. SIGNIFICANCE: Antiprogestin therapy induces immunogenic tumor cell death in PRA-overexpressing tumors, eliciting an adaptive immune memory response that protects mice from future tumor recurrence and increases sensitivity to PD-L1 blockade. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/5/1375/F1.large.jpg.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/inmunología , Animales , Antígeno B7-H1/antagonistas & inhibidores , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Humanos , Memoria Inmunológica/efectos de los fármacos , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos BALB C , Ratones Transgénicos , Mifepristona/farmacología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
3.
Medicina (B Aires) ; 79(Spec 6/1): 540-545, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31864223

RESUMEN

Seventy per cent of breast cancers are luminal carcinomas that express estrogen receptor alpha (ER). For several decades, its expression has been used as a therapeutic target in patients with breast cancer. These therapies are aimed at blocking ER or inhibiting ligand synthesis. The expression of progesterone receptors (PR) is evaluated as a prognostic factor together with ER. It has been shown that there are two predominant PR isoforms with different molecular weight, isoform A and isoform B, which are not distinguished by immunohistochemical techniques. The available evidence indicates that the PR isoform ratio may have both a prognostic and predictive value of the response to antiprogestin treatment. In luminal mammary carcinomas, androgen receptors (AR) are expressed in a high percentage and the AR/ER or AR/PR ratio could be a prognostic factor. In ER negative (-) tumors, AR expression is an indicator of poor prognosis and it is proposed that they may be susceptible to antiandrogen treatment. Finally, the expression of glucocorticoid receptors (GR) would be an indicator of good or bad prognosis in luminal or ER- tumors, respectively. In ER- tumors, metastases express higher levels of nuclear GR than primary tumors and therapies that block GR could improve the efficacy of chemotherapy. Given the crosstalk of pathways triggered by different hormone receptors, it is possible that in the future, a therapeutic scheme can be administered that contemplates the expression of ER, PR isoforms, AR and GR.


El 70 por ciento de los carcinomas mamarios son luminales y expresan receptores de estrógenos alfa (RE). Desde hace varias décadas, su expresión se utiliza como blanco terapéutico en pacientes con cáncer de mama. Estas terapias están dirigidas a bloquear el RE o a inhibir la síntesis del ligando. La expresión de receptores de progesterona (RP) se evalúa como factor pronóstico junto con los RE. Se ha comprobado que existen dos isoformas predominantes de RP de distinto peso molecular, isoforma A e isoforma B, que no se distinguen por técnicas de inmunohistoquímica. Las evidencias indican que la proporción de isoformas de RP podría tener tanto un valor pronóstico como predictivo de la respuesta a un tratamiento con antiprogestágenos. En tumores mamarios luminales, los receptores de andrógenos (RA) se expresan en un alto porcentaje y la proporción de RA/RE o RA/RP podría ser un factor pronóstico. En tumores RE-, la expresión de RA es indicador de mal pronóstico y se propone que serían susceptibles a un tratamiento con antiandrógenos. Por último, la expresión de receptores de glucocorticoides (RG) sería un indicador de buen o mal pronóstico en tumores luminales o RE-, respectivamente. En tumores RE-, las metástasis expresan mayores niveles de RG nuclear que los tumores primarios y las terapias que bloquean los RG podrían mejorar la eficacia de la quimioterapia. Dado los entrecruzamientos de vías gatilladas por distintos receptores hormonales es posible que en un futuro se pueda administrar un esquema terapéutico que contemple la expresión de RE, isoformas de RP, RA y RG.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Progesterona/metabolismo , Biomarcadores/metabolismo , Neoplasias de la Mama/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Pronóstico , Receptores Androgénicos/genética , Receptores de Estrógenos/genética , Receptores de Glucocorticoides/genética , Receptores de Progesterona/genética
4.
Biomed Opt Express ; 10(6): 2932-2941, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-31259063

RESUMEN

The photothermal effect is undergoing great interest due to advances in new photosensitizing materials and better-suited light sources, but studies are frequently hampered by the need to employ exogenous photothermal agents and expensive irradiation devices. Here we present a simple strategy based on direct NIR irradiation of the melanin pigment with a commercial 808-nm laser pointer. Proof-of-concept studies showed efficient photothermal effects on melanin in vitro and in vivo. After NIR irradiation, BALB/c mice bearing B16-F10 melanotic melanoma tumors revealed severe histopathological damage and massive necrosis in melanin-containing tumor tissue, while surrounding healthy tissues showed no damage. Therefore, the feasibility of this approach may allow implementing direct procedures for photothermal therapy of pigmented tumors.

5.
Biochim Biophys Acta Mol Cell Res ; 1866(8): 1338-1352, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30905597

RESUMEN

Galectin-8 (Gal-8), a 'tandem-repeat'-type galectin, has been described as a modulator of cellular functions including adhesion, spreading, growth arrest, apoptosis, pathogen recognition, autophagy, and immunomodulation. We have previously shown that activated leukocyte cell adhesion molecule (ALCAM), also known as CD166, serves as a receptor for endogenous Gal-8. ALCAM is a member of the immunoglobulin superfamily involved in cell-cell adhesion through homophilic (ALCAM-ALCAM) and heterophilic (i.e. ALCAM-CD6) interactions in different tissues. Here we investigated the physiologic relevance of ALCAM-Gal-8 association and glycosylation-dependent mechanisms governing these interactions. We found that silencing of ALCAM in MDA-MB-231 triple negative breast cancer cells decreases cell adhesion and migration onto Gal-8-coated surfaces in a glycan-dependent fashion. Remarkably, either Gal-8 or ALCAM silencing also disrupted cell-cell adhesion, and led to reduced tumor growth in a murine model of triple negative breast cancer. Moreover, structural characterization of endogenous ALCAM N-glycosylation showed abundant permissive structures for Gal-8 binding. Importantly, we also found that cell sialylation controls Gal-8-mediated cell adhesion. Altogether, these findings demonstrate a central role of either ALCAM or Gal-8 (or both) in controlling triple negative breast cancer.


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas Fetales/metabolismo , Galectinas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Antígenos CD/genética , Adhesión Celular/genética , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular Tumoral , Femenino , Proteínas Fetales/genética , Galectinas/genética , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteínas de Neoplasias/genética , Neoplasias de la Mama Triple Negativas/genética
6.
Lasers Med Sci ; 33(6): 1307-1315, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29549555

RESUMEN

The photothermal effect is one of the most promising photonic procedures currently under development to successfully treat several clinical disorders, none the least some kinds of cancer. At present, this field is undergoing a renewed interest due to advances in both photothermal materials and better-suited light sources. However, scientific studies in this area are sometimes hampered by the relative unavailability of state-of-art materials or the complexity of setting up a dedicated optical facility. Here, we present a simple and affordable approach to do research in the photothermal field that relies on a commercial NIR laser pointer and a readily available everyday pigment: China ink. A proof-of-concept study is presented in which mice bearing intradermal LM3 mammary adenocarcinoma tumors were successfully treated in vivo employing China ink and the laser pointer. TUNEL and Ki-67 post-treatment tissue assessment clearly indicates the deleterious action of the photothermal treatment on the tumor. Therefore, the feasibility of this simple approach has been demonstrated, which may inspire other groups to implement simple procedures to further explore the photothermal effect.


Asunto(s)
Hipertermia Inducida , Rayos Infrarrojos , Tinta , Rayos Láser , Neoplasias/terapia , Fototerapia , Animales , Apoptosis , Línea Celular Tumoral , China , Ratones , Neoplasias/patología
7.
Immunology ; 145(1): 82-93, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25438991

RESUMEN

The modulatory effects of solar UV radiation on the immune system have been widely studied. As the skin is the main target of UV radiation, our purpose was to compare the impact on skin innate immunity of two contrasting ways to be exposed to sunlight. Hairless mice were UV irradiated with a single high UV dose simulating a harmful exposure, or with repetitive low UV doses simulating short occasional daily exposures. Skin samples were taken at different times after UV irradiation to evaluate skin histology, inflammatory cell recruitment, epidermal T-cell population and the mitochondrial function of epidermal cells. The transcriptional profiles of pro-inflammatory cytokines, chemokines, antimicrobial peptides and Toll-like receptors were evaluated by RT-PCR and ELISA in tissue homogenates. Finally, a lymphangiography was performed to assess modification in the lymphatic vessel system. A single high UV dose produces a deep inflammatory state characterized by the production of pro-inflammatory cytokines and chemokines that, in turn, induces the recruitment of neutrophils and macrophages into the irradiated area. On the other hand, repetitive low UV doses drive the skin to a photo-induced alert state in which there is no sign of inflammation, but the epithelium undergoes changes in thickness, the lymphatic circulation increases, and the transcription of antimicrobial peptides is induced.


Asunto(s)
Inmunidad Innata/efectos de la radiación , Mediadores de Inflamación/inmunología , Piel/inmunología , Linfocitos T/inmunología , Rayos Ultravioleta/efectos adversos , Animales , Péptidos Catiónicos Antimicrobianos/inmunología , Quimiocinas/inmunología , Relación Dosis-Respuesta en la Radiación , Macrófagos/inmunología , Macrófagos/patología , Ratones , Infiltración Neutrófila/inmunología , Infiltración Neutrófila/efectos de la radiación , Neutrófilos/inmunología , Neutrófilos/patología , Piel/patología , Linfocitos T/patología , Factores de Tiempo , Receptores Toll-Like/inmunología
8.
Oncotarget ; 5(10): 3246-60, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24912774

RESUMEN

There is clinical and experimental evidence suggesting that antiprogestins might be used for the treatment of selected breast cancer patients. Our aim was to evaluate the effect of albumin-bound paclitaxel (Nab-paclitaxel) and pegylated doxorubicin liposomes (PEG-LD) in combination with mifepristone (MFP) in experimental breast cancer models expressing different ratios of progesterone receptor (PR) isoforms A and B. We used two antiprogestin-responsive (PRA>PRB) and two resistant (PRA

Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/metabolismo , Mifepristona/administración & dosificación , Receptores de Progesterona/metabolismo , Albúminas/administración & dosificación , Animales , Doxorrubicina/administración & dosificación , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Liposomas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas , Paclitaxel/administración & dosificación , Isoformas de Proteínas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Medicina (B Aires) ; 72(4): 315-20, 2012.
Artículo en Español | MEDLINE | ID: mdl-22892083

RESUMEN

In C4-HD murine mammary carcinomas and in human breast cancer T47D cells, we showed that medroxyprogesterone acetate (MPA) induces a nuclear physical association between estrogen receptor alpha (ERa) and progesterone receptors (PR). The blockade of ERa inhibits cell proliferation mediated by progestins. We hypothesized that this nuclear association between ERa/PR is necessary to trigger progestin-induced cell proliferation and tumor growth. We demonstrated that fulvestrant (FUL, ICI182.780) induced complete regression of C4-HD tumors growing with progestins. MPA treatment induced an early increase in both CCND1 and MYC expression in T47D cells. The blockade of ERa prevented the MPA-dependent transcription of both genes. Specific binding of PR/ERa was observed at the same MPA-sensitive regions at the CCND1 and MYC gene promoters after chromatin immunoprecipitation (ChIP) analysis. ICI inhibited binding of ERa to both gene regulatory sequences while PR binding was unaffected. The nuclear colocalization between both receptors in T47D cells was confirmed by: confocal microscopy, Duolink assays and co-immunoprecipitation assays. In breast cancer samples we also observed a nuclear interaction between both steroid receptors. Our results indicate that the presence of ERa interacting with activated PR at the CCND1 and MYC promoters is required to trigger progestin-induced gene transcription and cell proliferation in breast cancer cells.


Asunto(s)
Carcinoma/patología , Estradiol/análogos & derivados , Receptor alfa de Estrógeno/fisiología , Neoplasias Mamarias Experimentales/patología , Receptores de Progesterona/fisiología , Animales , Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/genética , Carcinoma/inducido químicamente , Carcinoma/tratamiento farmacológico , Proliferación Celular , Inmunoprecipitación de Cromatina , Ciclina D1/metabolismo , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/efectos de los fármacos , Femenino , Fulvestrant , Genes myc , Humanos , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Acetato de Medroxiprogesterona/farmacología , Murinae , Progestinas/metabolismo , Receptores de Progesterona/efectos de los fármacos , Transcripción Genética
10.
Cancer Res ; 72(9): 2416-27, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22396492

RESUMEN

Synthetic progesterone used in contraception drugs (progestins) can promote breast cancer growth, but the mechanisms involved are unknown. Moreover, it remains unclear whether cytoplasmic interactions between the progesterone receptor (PR) and estrogen receptor alpha (ERα) are required for PR activation. In this study, we used a murine progestin-dependent tumor to investigate the role of ERα in progestin-induced tumor cell proliferation. We found that treatment with the progestin medroxyprogesterone acetate (MPA) induced the expression and activation of ERα, as well as rapid nuclear colocalization of activated ERα with PR. Treatment with the pure antiestrogen fulvestrant to block ERα disrupted the interaction of ERα and PR in vitro and induced the regression of MPA-dependent tumor growth in vivo. ERα blockade also prevented an MPA-induced increase in CYCLIN D1 (CCND1) and MYC expression. Chromatin immunoprecipitation studies showed that MPA triggered binding of ERα and PR to the CCND1 and MYC promoters. Interestingly, blockade or RNAi-mediated silencing of ERα inhibited ERα, but not PR binding to both regulatory sequences, indicating that an interaction between ERα and PR at these sites is necessary for MPA-induced gene expression and cell proliferation. We confirmed that nuclear colocalization of both receptors also occurred in human breast cancer samples. Together, our findings argued that ERα-PR association on target gene promoters is essential for progestin-induced cell proliferation.


Asunto(s)
Neoplasias de la Mama/patología , Ciclina D1/genética , Receptor alfa de Estrógeno/metabolismo , Genes myc , Neoplasias Mamarias Experimentales/patología , Receptores de Progesterona/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Procesos de Crecimiento Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Estradiol/análogos & derivados , Estradiol/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Femenino , Fulvestrant , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Acetato de Medroxiprogesterona/farmacología , Ratones , Ratones Endogámicos BALB C , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Regiones Promotoras Genéticas , Receptores de Progesterona/genética
11.
Breast Cancer Res Treat ; 133(3): 997-1008, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22124578

RESUMEN

Fibroblast growth factor receptors (FGFRs) are tyrosine kinase receptors which have been implicated in breast cancer. The aim of this study was to evaluate FGFR-1, -2, -3, and -4 protein expressions in normal murine mammary gland development, and in murine and human breast carcinomas. Using immunohistochemistry and Western blot, we report a hormonal regulation of FGFR during postnatal mammary gland development. Progestin treatment of adult virgin mammary glands resulted in changes in localization of FGFR-3 from the cytoplasm to the nucleus, while treatment with 17-ß-estradiol induced changes in the expressions and/or localizations of FGFR-2 and -3. In murine mammary carcinomas showing different degrees of hormone dependence, we found progestin-induced increased expressions, mainly of FGFR-2 and -3. These receptors were constitutively activated in hormone-independent variants. We studied three luminal human breast cancer cell lines growing as xenografts, which particularly expressed FGFR-2 and -3, suggesting a correlation between hormonal status and FGFR expression. Most importantly, in breast cancer samples from 58 patients, we found a strong association (P < 0.01; Spearman correlation) between FGFR-2 and -3 expressions and a weaker correlation of each receptor with estrogen receptor expression. FGFR-4 correlated with c-erbB2 over expression. We conclude that FGFR-2 and -3 may be mechanistically linked and can be potential targets for treatment of estrogen receptor-positive breast cancer patients.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glándulas Mamarias Animales/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Adulto , Anciano , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Persona de Mediana Edad , Embarazo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Trasplante Heterólogo
12.
Infect Immun ; 79(9): 3619-32, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21730088

RESUMEN

Arthritis is one of the most common complications of human brucellosis, but its pathogenic mechanisms have not been elucidated. Fibroblast-like synoviocytes (FLS) are known to be central mediators of joint damage in inflammatory arthritides through the production of matrix metalloproteinases (MMPs) that degrade collagen and of cytokines and chemokines that mediate the recruitment and activation of leukocytes. In this study we show that Brucella abortus infects and replicates in human FLS (SW982 cell line) in vitro and that infection results in the production of MMP-2 and proinflammatory mediators (interleukin-6 [IL-6], IL-8, monocyte chemotactic protein 1 [MCP-1], and granulocyte-macrophage colony-stimulating factor [GM-CSF]). Culture supernatants from Brucella-infected FLS induced the migration of monocytes and neutrophils in vitro and also induced these cells to secrete MMP-9 in a GM-CSF- and IL-6-dependent fashion, respectively. Reciprocally, culture supernatants from Brucella-infected monocytes and neutrophils induced FLS to produce MMP-2 in a tumor necrosis factor alpha (TNF-α)-dependent fashion. The secretion of proinflammatory mediators and MMP-2 by FLS did not depend on bacterial viability, since it was also induced by heat-killed B. abortus (HKBA) and by a model Brucella lipoprotein (L-Omp19). These responses were mediated by the recognition of B. abortus antigens through Toll-like receptor 2. The intra-articular injection of HKBA or L-Omp19 into the knee joint of mice resulted in the local induction of the proinflammatory mediators MMP-2 and MMP-9 and in the generation of a mixed inflammatory infiltrate. These results suggest that FLS, and phagocytes recruited by them to the infection focus, may be involved in joint damage during brucellar arthritis through the production of MMPs and proinflammatory mediators.


Asunto(s)
Artritis Infecciosa/inmunología , Brucella abortus/inmunología , Brucelosis/inmunología , Articulaciones/microbiología , Articulaciones/patología , Metaloproteinasas de la Matriz/biosíntesis , Membrana Sinovial/inmunología , Animales , Antígenos Bacterianos/inmunología , Artritis Infecciosa/enzimología , Artritis Infecciosa/microbiología , Artritis Infecciosa/patología , Proteínas de la Membrana Bacteriana Externa/inmunología , Brucella abortus/crecimiento & desarrollo , Brucella abortus/patogenicidad , Brucelosis/enzimología , Brucelosis/microbiología , Brucelosis/patología , Línea Celular , Movimiento Celular/efectos de los fármacos , Quimiocinas/biosíntesis , Medios de Cultivo Condicionados , Citocinas/biosíntesis , Citocinas/metabolismo , Inducción Enzimática , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Articulación de la Rodilla/microbiología , Lipoproteínas/inmunología , Activación de Linfocitos , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Monocitos/fisiología , Neutrófilos/fisiología , Membrana Sinovial/citología , Membrana Sinovial/microbiología , Receptor Toll-Like 2/metabolismo
13.
Cancer Res ; 71(10): 3720-31, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21464042

RESUMEN

Fibroblast growth factor (FGF) receptor 2 (FGFR-2) polymorphisms have been associated with an increase in estrogen receptor and progesterone receptor (PR)-positive breast cancer risk; however, a clear mechanistic association between FGFR-2 and steroid hormone receptors remains elusive. In previous works, we have shown a cross talk between FGF2 and progestins in mouse mammary carcinomas. To investigate the mechanisms underlying these interactions and to validate our findings in a human setting, we have used T47D human breast cancer cells and human cancer tissue samples. We showed that medroxyprogesterone acetate (MPA) and FGF2 induced cell proliferation and activation of ERK, AKT, and STAT5 in T47D and in murine C4-HI cells. Nuclear interaction between PR, FGFR-2, and STAT5 after MPA and FGF2 treatment was also showed by confocal microscopy and immunoprecipitation. This effect was associated with increased transcription of PRE and/or GAS reporter genes, and of PR/STAT5-regulated genes and proteins. Two antiprogestins and the FGFR inhibitor PD173074, specifically blocked the effects induced by FGF2 or MPA respectively. The presence of PR/FGFR-2/STAT5 complexes bound to the PRE probe was corroborated by using NoShift transcription and chromatin immunoprecipitation of the MYC promoter. Additionally, we showed that T47D cells stably transfected with constitutively active FGFR-2 gave rise to invasive carcinomas when transplanted into NOD/SCID mice. Nuclear colocalization between PR and FGFR-2/STAT5 was also observed in human breast cancer tissues. This study represents the first demonstration of a nuclear interaction between FGFR-2 and STAT5, as PR coactivators at the DNA progesterone responsive elements, suggesting that FGFRs are valid therapeutic targets for human breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Progesterona/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Acetato de Medroxiprogesterona/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias
14.
Nitric Oxide ; 23(3): 234-41, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20633694

RESUMEN

Low O(2) levels in solid tumors are associated with increase in hypoxia-inducible factor 1alpha (HIF-1alpha). The present study examines functional changes involved in adaptation to hypoxia of the LMM3 mammary tumor cell line, using CoCl(2) as hypoxic mimetic. Our results showed that LMM3 cells were not only tolerant to 150 microM CoCl(2) but they can overgrowth in vitro respect to untreated cells. Hypoxia inhibited cell invasion, migration, MMP-9 activity and NO levels. Macrophage cytotoxicity augmented under hypoxia but was blunted by conditioned media from tumor cells. In vivo tumorigenicity of CoCl(2)-treated cells was greater than controls. Our results show stabilization of HIF-1alpha in LMM3 cells under CoCl(2) and functional changes associated with enhanced cell survival and growth but not with tumor dissemination.


Asunto(s)
Cobalto/farmacología , Hipoxia , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/biosíntesis , Relación Estructura-Actividad , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/biosíntesis
15.
BMC Cancer ; 10: 293, 2010 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-20553594

RESUMEN

BACKGROUND: Increasing evidence has underscored the role of carcinoma associated fibroblasts (CAF) in tumor growth. However, there are controversial data regarding the persistence of inoculated CAF within the tumors. We have developed a model in which murine metastatic ductal mammary carcinomas expressing estrogen and progesterone receptors transit through different stages of hormone dependency. Hormone dependent (HD) tumors grow only in the presence of progestins, whereas hormone independent (HI) variants grow without hormone supply. We demonstrated previously that CAF from HI tumors (CAF-HI) express high levels of FGF-2 and that FGF-2 induced HD tumor growth in vivo. Our main goal was to investigate whether inoculated CAF-HI combined with purified epithelial (EPI) HD cells can induce HD tumor growth. METHODS: Purified EPI cells of HD and HI tumors were inoculated alone, or together with CAF-HI, into female BALB/c mice and tumor growth was evaluated. In another set of experiments, purified EPI-HI alone or combined with CAF-HI or CAF-HI-GFP were inoculated into BALB/c or BALB/c-GFP mice. We assessed whether inoculated CAF-HI persisted within the tumors by analyzing inoculated or host CAF in frozen sections of tumors growing in BALB/c or BALB/c-GFP mice. The same model was used to evaluate early stages of tumor development and animals were euthanized at 2, 7, 12 and 17 days after EPI-HI or EPI-HI+CAF-HI inoculation. In angiogenesis studies, tumor vessels were quantified 5 days after intradermal inoculation. RESULTS: We found that admixed CAF-HI failed to induce epithelial HD tumor growth, but instead, enhanced HI tumor growth (p < 0.001). Moreover, inoculated CAF-HI did not persist within the tumors. Immunofluorescence studies showed that inoculated CAF-HI disappeared after 13 days. We studied the mechanisms by which CAF-HI increased HI tumor growth, and found a significant increase in angiogenesis (p < 0.05) in the co-injected mice at early time points. CONCLUSIONS: Inoculated CAF-HI do not persist within the tumor mass although they play a role during the first stages of tumor formation promoting angiogenesis. This angiogenic environment is unable to replace the hormone requirement of HD tumors that still need the hormone to recruit the stroma from the host.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Comunicación Celular , Proliferación Celular , Células Epiteliales/patología , Fibroblastos/patología , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/irrigación sanguínea , Carcinoma Ductal de Mama/inmunología , Carcinoma Ductal de Mama/metabolismo , Supervivencia Celular , Células Epiteliales/metabolismo , Femenino , Fibroblastos/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Trasplante de Neoplasias , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Factores de Tiempo , Carga Tumoral , Células Tumorales Cultivadas
16.
Breast Cancer Res ; 9(2): R22, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17341305

RESUMEN

INTRODUCTION: Medroxyprogesterone acetate (MPA) induces estrogen receptor (ER)-positive and progesterone receptor (PR)-positive ductal invasive mammary carcinomas in BALB/c mice. We sought to reproduce this MPA cancer model in C57BL/6 mice because of their widespread use in genetic engineering. Within this experimental setting, we studied the carcinogenic effects of MPA, the morphologic changes in mammary glands that are induced by MPA and progesterone, and the levels of ER and PR expression in MPA-treated and progesterone-treated mammary glands. Finally, we evaluated whether the differences found between BALB/c and C57BL/6 mouse strains were due to intrinsic differences in epithelial cells. METHODS: The carcinogenic effect of MPA was evaluated in C57BL/6 mice using protocols proven to be carcinogenic in BALB/c mice. In addition, BALB/c and C57BL/6 females were treated with progesterone or MPA for 1 or 2 months, and mammary glands were excised for histologic studies and for immunohistochemical and Western blot evaluation of ER and PR. Hormone levels were determined by radioimmunoassay. Isolated mammary epithelial cells were transplanted into cleared fat pads of 21-day-old female Swiss nu/nu mice or control congenic animals. RESULTS: MPA failed to induce mammary carcinomas or significant morphologic changes in the mammary glands of C57BL/6 mice. The expression of ER-alpha and PR isoform A in virgin mice was surprisingly much higher in BALB/c than in C57BL/6 mammary glands, and both receptors were downregulated in progestin-treated BALB/c mice (P < 0.05). PR isoform B levels were low in virgin control mice and increased after progestin treatment in both strains. ER-beta expression followed a similar trend. No differences in hormone levels were found between strains. Surprisingly, the transplantation of the epithelial mammary gland cells of both strains into the cleared fat pads of Swiss (nu/nu) mice abolished the mammary gland morphologic differences and the ER and PR differences between strains. CONCLUSION: C57BL/6 mammary glands are resistant to MPA-induced carcinogenesis and to hormone action. MPA and progesterone have different effects on mammary glands. Low ER-alpha and PR-A levels in untreated mammary glands may be associated with a low-risk breast cancer profile. Although we cannot at this time rule out the participation of other, untested factors, our findings implicate the stroma as playing a crucial role in the strain-specific differential hormone receptor expression and hormone responsiveness.


Asunto(s)
Receptor alfa de Estrógeno/biosíntesis , Neoplasias Mamarias Animales/inducido químicamente , Neoplasias Mamarias Animales/metabolismo , Receptores de Progesterona/biosíntesis , Animales , Carcinógenos , Células Epiteliales/metabolismo , Estradiol/metabolismo , Femenino , Hormonas/metabolismo , Acetato de Medroxiprogesterona , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Progesterona , Radioinmunoensayo
17.
Int J Oncol ; 27(4): 1053-9, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16142323

RESUMEN

Photodynamic therapy (PDT) is based on the cytotoxic effect induced by a photosensitizer in the presence of light and molecular oxygen, with production of reactive oxygen species which cause cell death and tumor destruction. Here we describe the response of the murine mammary adenocarcinoma, LM3, to repeated PDT treatments using the synthetic porphyrin derivative, meso-tetra (4-N-methylpyridinium) porphine (TMPyP). Intradermal LM3 tumors in BALB/c mice were left untreated, only treated with light, only injected with 0.9% NaCl solution, or with TMPyP alone (10 microg in 0.1 ml of 0.9% NaCl). For PDT, the intratumoral TMPyP injection was followed 1 h later by blue-red light irradiation for 50 min (80 mW/cm2 total dose: 240 J/cm2). In all cases, control and PDT treatments were performed on the depilated and glycerol-covered skin over the tumor of anesthetized mice and repeated four times (every two days). In a pilot experiment, no significant differences were found in the growth rate of untreated tumors (n=4) and tumors only treated with light (n=4), 0.9% NaCl (n=3) or TMPyP (n=3). PDT-treated tumors (n=3) showed transitory regression and growth delay. In a second approach, the average diameter (mean, mm +/- SEM) of control (drug alone, n=15) vs PDT tumors (n=17) was 2.13+/-0.11 vs 2.02+/-0.10 at day 0, and 4.00+/-0.17 vs 0.20+/-0.07 at day 9, p<0.0001. At day 37 the average diameter of tumors from control vs the PDT group was 10.98+/-0.59 vs 6.31+/-0.82, p<0.0001. PDT caused partial regression of tumors in one from a total of 17 mice, long-term regression in 15, and cure in one animal. Significant differences in the survival and tumor size at death were found between control and PDT-treated mice. Histopathological analysis of LM3 tumors one day after a unique PDT treatment showed extensive hemorrhage and necrotic areas. These results indicate the considerable potential of intratumoral injection of photosensitizers and repeated PDT protocols.


Asunto(s)
Neoplasias Mamarias Animales/terapia , Neoplasias Mamarias Experimentales/terapia , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacología , Animales , Proliferación Celular , Femenino , Glicerol/química , Hemorragia/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Químicos , Necrosis , Trasplante de Neoplasias , Oxígeno/metabolismo , Factores de Tiempo
18.
Carcinogenesis ; 26(6): 1055-63, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15774491

RESUMEN

In this paper we demonstrate, using the C7-2-HI metastatic transplantable ductal mammary tumor, that endocrine therapy can induce complete regression of spontaneous lymph node and lung metastases in a mouse model of breast cancer progression. This tumor expresses high levels of estrogen and progesterone receptors and shows a high incidence of early axillary lymph nodes and lung metastases; using this model we had previously shown complete tumor regression of subcutaneous implants. Interestingly, although the metastases showed a more differentiated histology as compared with the primary growth, they underwent complete regression when treated with estrogens or antiprogestins. This phenomenon was associated with sustained cytostasis and apoptosis accompanied by increases in p21 and p27 expression and early tissue remodeling. These results highlight the essential role of PR in regulating cell proliferation in this model as well as its possible use as therapeutic target.


Asunto(s)
Estradiol/uso terapéutico , Estrógenos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Mifepristona/uso terapéutico , Progestinas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Femenino , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Metástasis Linfática , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Mitosis/efectos de los fármacos , Receptores de Estrógenos/biosíntesis , Receptores de Progesterona/biosíntesis , Trasplante Heterólogo , Proteínas Supresoras de Tumor/biosíntesis
19.
Breast Cancer Res ; 7(6): R1111-21, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16457691

RESUMEN

INTRODUCTION: Previous data from our laboratory suggested that progesterone receptors (PRs) are involved in progestin-independent growth of mammary carcinomas. To investigate this possibility further, we studied the effects of PR antisense oligodeoxynucleotides (asPR) on in vivo tumor growth. METHOD: BALB/c mice with subcutaneous 25 mm2 mammary carcinomas expressing estrogen receptor-alpha and PR were either injected intraperitoneally with 1 mg asPR every 24 or 12 hours for 5-10 days, or subcutaneously with RU 486 (6.5 mg/kg body weight) every 24 hours. Control mice received vehicle or scPR. RESULTS: Significant inhibition of tumor growth as well as a significant decrease in bromodeoxyuridine uptake was observed in asPR-treated mice, which correlated with histological signs of regression and increased apoptosis. Mice treated with RU 486 experienced almost complete tumor regression. No differences were detected between vehicle-treated and scPR-treated mice. Anti-progestin-treated and asPR-treated mice were in a continuous estrous/meta-estrous state. Decreased phosphorylated extracellular signal-regulated kinase (ERK)1 and ERK2 levels and estrogen receptor-alpha expression were observed as late events in RU 486-treated and asPR-treated mice with regressing tumors. CONCLUSION: We demonstrate, for the first time, inhibition of tumor growth in vivo using asPR. Our results provide further evidence for a critical and hierarchical role of the PR pathway in mammary carcinomas.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Oligonucleótidos Antisentido/farmacología , Receptores de Progesterona/fisiología , Animales , Neoplasias de la Mama/fisiopatología , Receptor alfa de Estrógeno/biosíntesis , Femenino , Humanos , Infusiones Parenterales , Inyecciones Subcutáneas , Neoplasias Mamarias Experimentales , Ratones , Ratones Endogámicos BALB C , Oligonucleótidos Antisentido/administración & dosificación , Receptores de Progesterona/antagonistas & inhibidores
20.
Bol. Acad. Nac. Med. B.Aires ; 81(1): 67-82, ene.-jun. 2003. graf
Artículo en Español | LILACS | ID: lil-401449

RESUMEN

Carcinomas mamarios murinos, originalmente inducidos por acetato de medroxiprogesterona, con crecimiento autónomo y receptores de estrógenos y progesterona regresionan con estradiol (E2) o con antiprogestágenos. Con el objeto de analizar los mecanismos de la regresión tumoral, estudiamos las características morfológicas y la participación de los reguladores del ciclo celular tales como p21, p27, p53 y MDM2 mediante inmunohistoquímica utilizando dos tumores sensibles al E2 o a los antiprogestágenos y uno sensible al E2 y antiprogestágenos resistente. La regresión se asocia a disminución del parénquima tumoral con aumento del estroma, a un efecto antiproliferativo y proapoptótico y a la inducción de proteínas inhibitorias del ciclo celular tales como p21 y p27. Sin embargo el patrón de expresión de los reguladores del ciclo celular varió. En los tumores sensibles a ambos tratamientos aumentó p21 y p27, los valores basales de p53 fueron altos y los de MDM2 bajos. En el tumor sensible sólo a E2 aumentó únicamente p27 y p21 permaneció en valores bajos acompañados por altos niveles basales de p53 y MDM2. Estos hallazgos sugieren que p21 podría ser esencial para la acción de los antiprogestágenos y que alteraciones en la vía p21/p53 podrían participar en la resistencia al tratamiento hormonal.


Asunto(s)
Animales , Femenino , Estrógenos/administración & dosificación , Estrógenos/uso terapéutico , Terapia de Reemplazo de Hormonas , Neoplasias Mamarias Experimentales , Inducción de Remisión , Apoptosis , Estradiol/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Mitosis , Medroxiprogesterona/toxicidad , Proteína Oncogénica p21(ras) , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Progesterona/antagonistas & inhibidores , Proteína p53 Supresora de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...