Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Intervalo de año de publicación
1.
Int J Cancer ; 150(9): 1481-1496, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-34935137

RESUMEN

Progesterone receptors (PRs) ligands are being tested in luminal breast cancer. There are mainly two PR isoforms, PRA and PRB, and their ratio (PRA/PRB) may be predictive of antiprogestin response. Our aim was to investigate: the impact of the PR isoform ratio on metastatic behaviour, the PR isoform ratio in paired primary tumours and lymph node metastases (LNM) and, the effect of antiprogestin/progestins on metastatic growth. Using murine and human metastatic models, we demonstrated that tumours with PRB > PRA (PRB-H) have a higher proliferation index but less metastatic ability than those with PRA > PRB (PRA-H). Antiprogestins and progestins inhibited metastatic burden in PRA-H and PRB-H models, respectively. In breast cancer samples, LNM retained the same PRA/PRB ratio as their matched primary tumours. Moreover, PRA-H LNM expressed higher total PR levels than the primary tumours. The expression of NDRG1, a metastasis suppressor protein, was higher in PRB-H compared to PRA-H tumours and was inversely regulated by antiprogestins/progestins. The binding of the corepressor SMRT at the progesterone responsive elements of the NDRG1 regulatory sequences, together with PRA, impeded its expression in PRA-H cells. Antiprogestins modulate the interplay between SMRT and AIB1 recruitment in PRA-H or PRB-H contexts regulating NDRG1 expression and thus, metastasis. In conclusion, we provide a mechanistic interpretation to explain the differential role of PR isoforms in metastatic growth and highlight the therapeutic benefit of using antiprogestins in PRA-H tumours. The therapeutic effect of progestins in PRB-H tumours is suggested.


Asunto(s)
Neoplasias de la Mama , Proteínas de Ciclo Celular , Péptidos y Proteínas de Señalización Intracelular , Receptores de Progesterona , Animales , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Metástasis de la Neoplasia , Progesterona/farmacología , Progestinas/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/metabolismo
2.
J Cell Biochem ; 119(7): 5402-5412, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29363820

RESUMEN

Conservative treatment for invasive bladder cancer (BC) involves a complete transurethral tumor resection combined with chemotherapy (CT) and radiotherapy (RT). The major obstacles of chemo-radiotherapy are the addition of the toxicities of RT and CT, and the recurrence due to RT and CT resistances. The flavonoid Silybin (Sb) inhibits pathways involved in cell survival and resistance mechanisms, therefore the purpose of this paper was to study in vitro and in vivo, the ability of Sb to improve the response to RT, in two murine BC cell lines, with different levels of invasiveness, placing emphasis on radio-sensitivity, and pathways involved in radio-resistance and survival. In vitro, Sb radio-sensitized murine invasive cells through the inhibition of RT-induced NF-κB and PI3K pathways, and the increase of oxidative stress, while non-invasive cells did not show to be sensitized. In vivo, Sb improved RT-response and overall survival in invasive murine tumors. As Sb is already being tested in clinical trials for other urological cancers and it improves RT-response in invasive BC, these results could have translational relevance, supporting further research.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Rayos gamma , Fármacos Sensibilizantes a Radiaciones/farmacología , Silibina/farmacología , Neoplasias de la Vejiga Urinaria/radioterapia , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología
3.
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
4.
Infect Immun ; 84(2): 598-606, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26667834

RESUMEN

The liver is frequently affected in patients with active brucellosis. In the present study, we identified a virulence factor involved in the modulation of hepatic stellate cell function and consequent fibrosis during Brucella abortus infection. This study assessed the role of BPE005 protein from B. abortus in the fibrotic phenotype induced on hepatic stellate cells during B. abortus infection in vitro and in vivo. We demonstrated that the fibrotic phenotype induced by B. abortus on hepatic stellate (LX-2) cells was dependent on BPE005, a protein associated with the type IV secretion system (T4SS) VirB from B. abortus. Our results indicated that B. abortus inhibits matrix metalloproteinase 9 (MMP-9) secretion through the activity of the BPE005-secreted protein and induces concomitant collagen deposition by LX-2 cells. BPE005 is a small protein containing a cyclic nucleotide monophosphate binding domain (cNMP) that modulates the LX-2 cell phenotype through a mechanism that is dependent on the cyclic AMP (cAMP)/protein kinase A (PKA) signaling pathway. Altogether, these results indicate that B. abortus tilts LX-2 cells to a profibrogenic phenotype employing a functional T4SS and the secreted BPE005 protein through a mechanism that involves the cAMP and PKA signaling pathway.


Asunto(s)
Proteínas Bacterianas/química , Brucella abortus/metabolismo , Colágeno/metabolismo , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/microbiología , Hígado/patología , Metaloproteinasa 9 de la Matriz/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Brucella abortus/química , Brucella abortus/genética , Brucella abortus/patogenicidad , Brucelosis/microbiología , Brucelosis/patología , Línea Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Femenino , Fibrosis , Regulación Enzimológica de la Expresión Génica , Células Estrelladas Hepáticas/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Fenotipo , Transducción de Señal , Sistemas de Secreción Tipo IV , Factores de Virulencia
5.
Tumour Biol ; 37(10): 13637-13647, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27473081

RESUMEN

Mycosis fungoides is the most common type of primary cutaneous T cell lymphoma. We have evaluated CDKN2A losses and MYC gains/amplifications by FISH analysis, as well as expression of miR-155 and members of the oncogenic cluster miR-17-92 (miR17, miR18a, miR19b, and miR92a) in MF patients with advanced disease. Formalin-fixed paraffin-embedded skin biopsies from 36 patients at diagnosis, 16 with tumoral MF (T-MF), 13 in histological transformation to a large T cell lymphoma (TR-MF), and 7 cases with folliculotropic variant (F-MF), were studied. Twenty cases showed genomic alterations (GAs): 8 (40 %) had CDKN2A deletion, 7 (35 %) showed MYC gain, and 5 (25 %) exhibited both alterations. GAs were more frequently observed in F-MF (p = 0.004) and TR-MF (p = 0.0001) than T-MF. GAs were significantly higher in cases presenting lesions in head, neck, and lower extremities compared to those observed in trunk and upper extremities (p = 0.03), when ≥25 % neoplastic cells were CD30 positive (p = 0.016) as well as in cases with higher Ki-67 proliferation index (p = 0.003). Patients with GAs showed bad response to treatment (p = 0.02) and short survival (p = 0.04). Furthermore, MF patients showed higher miRNA expression compared to controls (p ≤ 0.0223). T-MF showed higher miR17 and miR-18a expression compared to F-MF and TR-MF (p ≤ 0.0387) while miR19b, miR92a, and miR-155 showed increased levels in F-MF and TR-MF with respect to T-MF (p ≤ 0.0360). Increased expression of miR17 and miR19b in GA group compared to cases without alterations (p ≥ 0.0307) was also detected. Our results add new information about genomic imbalances in MF patients, particularly in F-MF, and extend the present view of miRNA deregulation in this disease.


Asunto(s)
Biomarcadores de Tumor/genética , Inestabilidad Genómica , Linfoma Cutáneo de Células T/genética , MicroARNs/genética , Micosis Fungoide/genética , Neoplasias Cutáneas/genética , Transcriptoma , Adulto , Anciano , Anciano de 80 o más Años , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 9/genética , Femenino , Estudios de Seguimiento , Genómica/métodos , Humanos , Hibridación Fluorescente in Situ , Linfoma Cutáneo de Células T/patología , Masculino , Persona de Mediana Edad , Micosis Fungoide/patología , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Cutáneas/patología
6.
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
7.
Int J Med Microbiol ; 305(8): 910-7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26456732

RESUMEN

Shiga toxin-producing Escherichia coli (STEC) is a food-borne pathogen that causes hemorrhagic colitis. Under some circumstances, Shiga toxin (Stx) produced within the intestinal tract enters the bloodstream, leading to systemic complications that may cause the potentially fatal hemolytic-uremic syndrome (HUS). Despite STEC human infection is characterized by acute inflammation of the colonic mucosa, little is known regarding the role of proinflammatory mediators like cysteine leukotrienes (cysLTs) in this pathology. Thus, the aim of this work was to analyze whether leukotriene C4 (LTC4) influences STEC pathogenesis in mice. We report that exogenous LTC4 pretreatment severely affected the outcome of STEC gastrointestinal infection. LTC4-pretreated (LTC4+) and STEC-infected (STEC+) mice showed an increased intestinal damage by histological studies, and a decreased survival compared to LTC4-non-pretreated (LTC4-) and STEC+ mice. LTC4+/STEC+ mice that died after the infection displayed neutrophilia and high urea levels, indicating that the cause of death was related to Stx2-toxicity. Despite the differences observed in the survival between LTC4+ and LTC4- mice after STEC infection, both groups showed the same survival after Stx2-intravenous inoculation. In addition, LTC4 pretreatment increased the permeability of mucosal intestinal barrier, as assessed by FITC-dextran absorption experiments. Altogether these results suggest that LTC4 detrimental effect on STEC infection is related to the increased passage of pathogenic factors to the bloodstream. Finally, we showed that STEC infection per se increases the endogenous LTC4 levels in the gut, suggesting that this inflammatory mediator plays a role in the pathogenicity of STEC infection in mice, mainly by disrupting the mucosal epithelial barrier.


Asunto(s)
Susceptibilidad a Enfermedades , Infecciones por Escherichia coli/complicaciones , Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/patología , Leucotrieno C4/metabolismo , Escherichia coli Shiga-Toxigénica/patogenicidad , Animales , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Intestinos/patología , Ratones Endogámicos BALB C , Análisis de Supervivencia
8.
Am J Pathol ; 183(6): 1918-1927, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24113459

RESUMEN

In patients with active brucellosis, the liver is frequently affected by histopathologic lesions, such as granulomas, inflammatory infiltrations, and parenchymal necrosis. Herein, we examine some potential mechanisms of liver damage in brucellosis. We demonstrate that Brucella abortus infection inhibits matrix metalloproteinase-9 (MMP-9) secretion and induces collagen deposition and tissue inhibitor of matrix metalloproteinase-1 secretion induced by hepatic stellate cells (LX-2). These phenomena depend on transforming growth factor-ß1 induction. In contrast, supernatants from B. abortus-infected hepatocytes and monocytes induce MMP-9 secretion and inhibit collagen deposition in hepatic stellate cells. Yet, if LX-2 cells are infected with B. abortus, the capacity of supernatants from B. abortus-infected hepatocytes and monocytes to induce MMP-9 secretion and inhibit collagen deposition is abrogated. These results indicate that depending on the balance between interacting cells and cytokines of the surrounding milieu, the response of LX-2 cells could be turned into an inflammatory or fibrogenic phenotype. Livers from mice infected with B. abortus displayed a fibrogenic phenotype with patches of collagen deposition and transforming growth factor-ß1 induction. This study provides potential mechanisms of liver immune response induced by B. abortus-infected hepatic stellate cells. In addition, these results demonstrate that the cross talk of these cells with hepatocytes and macrophages implements a series of interactions that may contribute to explaining some of mechanisms of liver damage observed in human brucellosis.


Asunto(s)
Brucella abortus , Brucelosis , Colágeno/metabolismo , Regulación Enzimológica de la Expresión Génica , Células Estrelladas Hepáticas , Cirrosis Hepática , Metaloproteinasa 9 de la Matriz/biosíntesis , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Brucelosis/metabolismo , Brucelosis/patología , Línea Celular , Regulación hacia Abajo , Femenino , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos BALB C
9.
Am J Pathol ; 180(3): 1040-1048, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22203055

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli is the main etiological agent that causes hemolytic uremic syndrome (HUS), a microangiopathic disease characterized by hemolytic anemia, thrombocytopenia, and acute renal failure. Although direct cytotoxic effects on endothelial cells by Stx are the primary pathogenic event, there is evidence that indicates the inflammatory response mediated by polymorphonuclear neutrophils and monocytes as the key event during HUS development. Because the chemokine receptor CCR1 participates in the pathogenesis of several renal diseases by orchestrating myeloid cell kidney infiltration, we specifically addressed the contribution of CCR1 in a murine model of HUS. We showed that Stx type 2-treated CCR1(-/-) mice have an increased survival rate associated with less functional and histological renal damage compared with control mice. Stx type 2-triggered neutrophilia and monocytosis and polymorphonuclear neutrophil and monocyte renal infiltration were significantly reduced and delayed in CCR1(-/-) mice compared with control mice. In addition, the increase of the inflammatory cytokines (tumor necrosis factor-α and IL-6) in plasma was delayed in CCR1(-/-) mice compared with control mice. These data demonstrate that CCR1 participates in cell recruitment to the kidney and amplification of the inflammatory response that contributes to HUS development. Blockade of CCR1 could be important to the design of future therapies to restrain the inflammatory response involved in the development of HUS.


Asunto(s)
Síndrome Hemolítico-Urémico/inducido químicamente , Receptores CCR1/fisiología , Toxina Shiga II/toxicidad , Animales , Médula Ósea/patología , Creatina/metabolismo , Síndrome Hemolítico-Urémico/patología , Interleucina-6/metabolismo , Túbulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Células Mieloides/fisiología , Receptores CCR1/deficiencia , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/metabolismo , Urea/metabolismo
10.
Cancer Chemother Pharmacol ; 92(6): 485-499, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37725114

RESUMEN

PURPOSE: Globally breast cancer accounts for 24.5% in incidence and 15.5% in cancer deaths in women. The triple-negative subtype lacks any specific therapy and is treated with chemotherapy, resulting in significant side-effects. We aimed to investigate if the dose of chemotherapeutic drugs could be diminished by co-administering it with the ß2-agonist salbutamol. METHODS: Cell proliferation was measured by thymidine incorporation; gene expression, by real-time PCR and protein phosphorylation by WB. Apoptosis was assessed by acridine orange / ethidium bromide and TUNEL tests. Public patient databases were consulted. Cells were inoculated to nude mice and their growth assessed. RESULTS: The ß2-agonist salbutamol synergizes in MDA-MB-231 cells in vitro with paclitaxel and doxorubicin on cell proliferation through ADRB2 receptors, while the ß-blocker propranolol does not. The expression of this receptor was assessed in patient databases and other cell lines. Triple negative samples had the lowest expression. Salbutamol and paclitaxel decreased MDA-MB-231 cell proliferation while their combination further inhibited it. The pathways involved were analyzed. When these cells were inoculated to nude mice, paclitaxel and salbutamol inhibited tumor growth. The combined effect was significantly greater. Paclitaxel increased the expression of MDR1 while salbutamol partially reversed this increase. CONCLUSION: While the effect of salbutamol was mainly on cell proliferation, suboptimal concentrations of paclitaxel provoked a very important enhancement of apoptosis. The latter enhanced transporter proteins as MDR1, whose expression were diminished by salbutamol. The expression of ADRB2 should be assessed in the biopsy or tumor to eventually select patients that could benefit from salbutamol repurposing.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama Triple Negativas , Animales , Ratones , Humanos , Femenino , Paclitaxel , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Ratones Desnudos , Albuterol/farmacología , Albuterol/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Propranolol , Agonistas Adrenérgicos/farmacología , Agonistas Adrenérgicos/uso terapéutico , Apoptosis
11.
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
12.
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
13.
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
14.
Front Immunol ; 13: 999852, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275674

RESUMEN

Histamine (HA) is a potent mediator that plays a central role in inflammation and allergy, acting through four G-protein-coupled receptors (i.e. H1-H4). HA is an accepted promoter of type 2 immunity in CD4+ T cells during hypersensitivity. Previously, we demonstrated that HA can promote antigen cross-presentation, inducing the activation of antigen-specific CD8+ T cells in an asthmatic murine model. Non-classical CD8+ T-cell profiles, such as Tc2 or Tc17, are associated with allergic disease persistence and chronicity. In this paper, we focus on the role of the H3 receptor (H3R) and the H4 receptor (H4R) in the development of allergic contact dermatitis. We were able to show that induction of the type 2 profiles associated with interleukin 13 production, both by CD4 and CD8 lymphocytes, depend on the interaction of HA with H3R and H4R. Blocking both receptors using the selective H3/H4 receptor antagonist thioperamide or the selective H4R ligand JNJ777120 reduces the inflammatory response, inducing an immunosuppressive profile associated with the increased proportion of FOXp3+ regulatory T lymphocytes and CD11b+Gr-1+ myeloid suppressor cells. Interestingly, in dendritic cells, only H4R blockade, and not H3R blockade, is capable of modulating most of the inflammatory effects observed in our model.


Asunto(s)
Dermatitis Alérgica por Contacto , Histamina , Ratones , Animales , Receptores Histamínicos H4 , Linfocitos T CD8-positivos , Ligandos , Interleucina-13 , Receptores Histamínicos , Receptores Acoplados a Proteínas G , Factores de Transcripción Forkhead
15.
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
16.
Sci Rep ; 11(1): 23820, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34893702

RESUMEN

Photodynamic therapy (PDT) is an anticancer treatment involving administration of a tumour-localizing photosensitizer, followed by activation by light of a suitable wavelength. In previous work, we showed that the natural anthraquinone (AQ) Parietin (PTN), was a promising photosensitizer for photodynamic therapy of leukemic cells in vitro. The present work aimed to analyze the photosensitizing ability of PTN in the mammary carcinoma LM2 cells in vitro and in vivo in a model of subcutaneously implanted tumours. Photodynamic therapy mediated by parietin (PTN-PDT) (PTN 30 µM, 1 h and 1.78 J/cm2 of blue light) impaired cell growth and migration of LM2 cells in vitro. PTN per se induced a significant decrease in cell migration, and it was even more marked after illumination (migration index was 0.65 for PTN and 0.30 for PTN-PDT, *p < 0.0001, ANOVA test followed by Tukey's multiple comparisons test), suggesting that both PTN and PTN-PDT would be potential inhibitors of metastasis. Fluorescence microscopy observation indicated cytoplasmic localization of the AQ and no fluorescence at all was recorded in the nuclei. When PTN (1.96 mg) dissolved in dimethyl sulfoxide was topically applied on the skin of mice subcutaneously implanted with LM2 cells, PTN orange fluorescence was strongly noticed in the stratum corneum and also in the inner layers of the tumour up to approximately 5 mm. After illumination with 12.74 J/cm2 of blue light, one PDT dose at day 1, induced a significant tumour growth delay at day 3, which was not maintained in time. Therefore, we administered a second PTN-PDT boost on day 3. Under these conditions, the delay of tumour growth was 28% both on days 3 and 4 of the experiment (*p < 0.05 control vs. PTN-PDT, two-way ANOVA, followed by Sidak's multiple comparisons test). Histology of tumours revealed massive tumour necrosis up to 4 mm of depth. Intriguingly, a superficial area of viable tumour in the 1 mm superficial area, and a quite conserved intact skin was evidenced. We hypothesize that this may be due to PTN aggregation in contact with the skin and tumour milieu of the most superficial tumour layers, thus avoiding its photochemical properties. On the other hand, normal skin treated with PTN-PDT exhibited slight histological changes. These preliminary findings encourage further studies of natural AQs administered in different vehicles, for topical treatment of cutaneous malignancies.


Asunto(s)
Antraquinonas/farmacología , Emodina/farmacología , Luz , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Neoplasias Cutáneas/terapia , Animales , Antraquinonas/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Emodina/química , Femenino , Ratones , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/metabolismo , Resultado del Tratamiento , Células Tumorales Cultivadas
17.
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
18.
Breast Cancer Res Treat ; 123(3): 709-24, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20012353

RESUMEN

To evaluate the extent to which each estrogen receptor (ER) subtype contributes to the stimulation or to the inhibition of mammary tumor growth, we evaluated the effects of specific agonists in MC4-L2 cells, which are stimulated by 17ß-estradiol (E(2)), and in mammary carcinomas of the MPA mouse breast cancer model, which are inhibited by E(2). Both express ERα and ERß. In MC4-L2 cells, 4,4',4"-(4-propyl-(1H)-pyrazole-1,3,5-triyl)trisphenol (PPT; ERα agonist) and (4-hydroxy-phenyl)-propionitrile (DPN; ERß agonist) stimulated cell proliferation, whereas the opposite occurred in C4-HI primary cultures. The inhibitory effect was associated with a decrease in ERα and cyclin D1 expression and an increase in progesterone receptor (PR) expression as well as in the Bax/Bcl-xl ratio. In vivo, mice carrying C4-HI or 32-2-HI tumors were treated with E(2), PPT or DPN (3 mg/kg/day) or with vehicle. PPT and DPN inhibited tumor size, as did E(2), during the first 72 h. After a few days, DPN-treated tumors started to grow again, while PPT-treated tumors remained quiescent for a longer period of time. A pronounced decrease in the mitotic index and an increase in the apoptotic index was associated with tumor regression. All treated tumors showed: (a) an increase in integrin α6 and Bax expression, (b) an increased stromal laminin redistribution, and (c) a decrease in ERα, Bcl-xl and Bcl-2 expression (P < 0.001). Apoptosis-inducing factor (Aif) expression was increased in DPN-treated tumors, while active caspase 9 was up-regulated in PPT-treated mice, demonstrating the involvement of the intrinsic apoptotic pathway in estrogen-induced regression in this model. In conclusion, our data indicate that although there may be some preferences for activation pathways by the different agonists, the stimulatory or inhibitory effects triggered by estrogens are cell-context dependent rather than ER isoform dependent.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Nitrilos/farmacología , Fenoles/farmacología , Pirazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Caspasa 9 , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Ratones , Ratones Endogámicos BALB C , Receptores de Progesterona/metabolismo , Factores de Tiempo , Carga Tumoral , Células Tumorales Cultivadas , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA