Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Life Sci ; 322: 121652, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37011871

ABSTRACT

AIMS: In white adipose tissue (WAT) the cell cycle regulators CDK4 and CDK6 (CDK4/6) promote adipogenesis and maintain the adipocyte mature state. Here we aimed to investigate their role in the Ucp1-mediated thermogenesis of WAT depots and in the biogenesis of beige adipocytes. MAIN METHODS: We treated mice with the CDK4/6 inhibitor palbociclib at room temperature (RT) or cold and analyzed thermogenic markers in the epididymal (abdominal) and inguinal (subcutaneous) WAT depots. We also assessed the effect of in vivo palbociclib-treatment on the percentage of beige precursors in the stroma vascular fraction (SVF), and on its beige adipogenic potential. Finally, we treated SVFs and mature adipocytes from WAT depots with palbociclib in vitro to study the role of CDK4/6 in beige adipocytes biogenesis. KEY FINDINGS: In vivo CDK4/6 inhibition downregulated thermogenesis at RT and impaired cold-induced browning of both WAT depots. It also reduced the percentage of beige precursors and beige adipogenic potential of the SVF upon differentiation. A similar result was observed with direct CDK4/6 inhibition in the SVF of control mice in vitro. Importantly, CDK4/6 inhibition also downregulated the thermogenic program of beige differentiated- and depots-derived adipocytes. SIGNIFICANCE: CDK4/6 modulate Ucp1-mediated thermogenesis of WAT depots in basal and cold-stressing conditions controlling beige adipocytes biogenesis by adipogenesis and transdifferentiation. This shows a pivotal role of CDK4/6 in WAT browning that could be applied to fight obesity or browning-associated hypermetabolic conditions such as cancer cachexia.


Subject(s)
Adipocytes , Adipose Tissue, White , Animals , Mice , Adipose Tissue, White/metabolism , Adipocytes/metabolism , Cell Differentiation , Adipogenesis , Thermogenesis , Adipose Tissue, Brown/metabolism , Uncoupling Protein 1/metabolism
2.
Sci Rep ; 13(1): 2710, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36792625

ABSTRACT

Resistance to therapy remains a major obstacle in cancer management. Although treatment with hormone and CDK4/6 inhibitors is successful in luminal breast cancer, resistance to these treatments is frequent, highlighting the need for novel therapeutic strategies to delay disease progression and improve patient survival. Here, we assessed the mechanisms of acquired resistance using T47D and MCF-7 tamoxifen- and palbociclib-resistant cell-line variants in culture and as xenografts, and patient-derived cells (PDCs) obtained from sensitive or resistant patient-derived xenografts (PDXs). In these models, we analyzed the effect of specific kinase inhibitors on survival, signaling and cellular aggressiveness. Our results revealed that mTOR inhibition is more effective than PI3K inhibition in overcoming resistance, irrespective of PIK3CA mutation status, by decreasing cell proliferation and tumor growth, as well as reducing cell migration and stemness. Moreover, a combination of mTOR and CDK4/6 inhibitors may prevent pathway reactivation downstream of PI3K, interfering with the survival of resistant cells and consequent tumor escape. In conclusion, we highlight the benefits of incorporating mTOR inhibitors into the current therapy in ER + breast cancer. This alternative therapeutic strategy not only enhances the antitumor response but may also delay the emergence of resistance and tumor recurrence.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Cell Line, Tumor , Neoplasm Recurrence, Local , TOR Serine-Threonine Kinases/metabolism , Hormones/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6
3.
Clin Cancer Res ; 29(5): 866-877, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36269797

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Mifepristone , Humans , Female , Mifepristone/pharmacology , Mifepristone/therapeutic use , Receptors, Progesterone/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Proteomics , Ki-67 Antigen , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
J Mammary Gland Biol Neoplasia ; 26(3): 227-234, 2021 09.
Article in English | MEDLINE | ID: mdl-34642841

ABSTRACT

The first Buenos Aires Breast Cancer Symposium (BA-BCS) was held in a virtual format, between the 17th and the 21st of May 2021. The main goal of the meeting was to facilitate the interaction among physicians and basic researchers from South America and with peers from the rest of the world. To embrace their different interests and concerns, the congress included not only talks on basic, translational and clinical research, but also round tables to discuss diagnostic methods, research financing and biobank management, as well as virtual poster sessions in which the youngest fellows presented their recent findings. This report provides a brief overview of the talks delivered during the meeting, which addressed a wide variety of vital issues for breast cancer research mostly focused on the accurate diagnosis, prevention and treatment of this illness. The presentations included a wide spectrum of themes including hormone receptors and the relevance of their mutations, immunotherapy, cancer stem cells, mouse models, environmental hazards, genetics and epigenetics, local and systemic therapies, liquid biopsies, the metastatic cascade, therapy resistance and dormancy, among others.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Clinical Trials as Topic , Translational Research, Biomedical , Argentina , Female , Humans , International Cooperation , Interprofessional Relations
5.
Cancer Res ; 81(5): 1375-1387, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33268529

ABSTRACT

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.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Cell Death/drug effects , Cell Death/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Female , Humans , Immunologic Memory/drug effects , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Transgenic , Mifepristone/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
6.
Int J Cancer ; 145(7): 1874-1888, 2019 10 01.
Article in English | MEDLINE | ID: mdl-30843188

ABSTRACT

Progression to hormone-independent growth leading to endocrine therapy resistance occurs in a high proportion of patients with estrogen receptor alpha (ERα) and progesterone receptors (PR) positive breast cancer. We and others have previously shown that estrogen- and progestin-induced tumor growth requires ERα and PR interaction at their target genes. Here, we show that fibroblast growth factor 2 (FGF2)-induces cell proliferation and tumor growth through hormone-independent ERα and PR activation and their interaction at the MYC enhancer and proximal promoter. MYC inhibitors, antiestrogens or antiprogestins reverted FGF2-induced effects. LC-MS/MS identified 700 canonical proteins recruited to MYC regulatory sequences after FGF2 stimulation, 397 of which required active ERα (ERα-dependent). We identified ERα-dependent proteins regulating transcription that, after FGF2 treatment, were recruited to the enhancer as well as proteins involved in transcription initiation that were recruited to the proximal promoter. Also, among the ERα-dependent and independent proteins detected at both sites, PR isoforms A and B as well as the novel protein product PRBΔ4 were found. PRBΔ4 lacks the hormone-binding domain and was able to induce reporter gene expression from estrogen-regulated elements and to increase cell proliferation when cells were stimulated with FGF2 but not by progestins. Analysis of the Cancer Genome Atlas data set revealed that PRBΔ4 expression is associated with worse overall survival in luminal breast cancer patients. This discovery provides a new mechanism by which growth factor signaling can engage nonclassical hormone receptor isoforms such as PRBΔ4, which interacts with growth-factor activated ERα and PR to stimulate MYC gene expression and hence progression to endocrine resistance.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Fibroblast Growth Factor 2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Receptors, Progesterone/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Prognosis , Promoter Regions, Genetic , Protein Interaction Maps , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Progesterone/genetics , Survival Analysis , Xenograft Model Antitumor Assays
7.
Eur J Med Genet ; 62(11): 103571, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30414531

ABSTRACT

Malformations of cortical development are a frequent cause of drug-resistant Epilepsy and developmental delay. Hemimegalencephaly is a Malformation of cortical development characterized by enlargement of all or a part of one cerebral hemisphere. Germline and somatic mutation in genes belonging to the Mammalian Target of Rapamycin (mTOR) pathway has been identified in patients suffering from epilepsy secondary to Hemimegalencephaly and focal cortical dysplasia. We present here a patient suffering from severe neonatal Epilepsy since 3 h of life secondary to Hemimegalencephaly, requiring an anatomic hemispherectomy surgical procedure for seizure control, where by means of next-generation sequencing at an ultra-high depth coverage, we were able to identify a novel somatic mutation in the RHEB gene (NM_005614: c.119A > T: p. Glu40Val). The histopathological diagnosis was Cortical Dysplasia type IIB determined by the presence of dysmorphic neurons of variable size with nuclear alteration and balloon cells in the context of Hemimegalencephaly, which are similar to that have been demonstrated in hyperactivating RHEB models. This is the first report of a somatic mutation in RHEB gene in a patient suffering from Epilepsy secondary to Hemimegalencephaly. It highlights different current topics in the fields of genetics of Malformations of cortical development: a-somatic mosaicism is not uncommon in these neurodevelopmental disorders; b-the molecular diagnostic approach should involve the use of state-of-the-art methods and the sampling of different tissues; c-new findings might facilitate therapeutics discoveries while providing an improved understanding of normal brain development.


Subject(s)
Drug Resistant Epilepsy/genetics , Hemimegalencephaly/genetics , Malformations of Cortical Development/genetics , Ras Homolog Enriched in Brain Protein/genetics , Drug Resistant Epilepsy/pathology , Female , Hemimegalencephaly/pathology , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Malformations of Cortical Development/pathology , Mutation , TOR Serine-Threonine Kinases/genetics
8.
Sci Rep ; 7: 44244, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28287129

ABSTRACT

The purpose of this study was to elucidate the mechanisms associated with the specific effects of AKT1 and AKT2 isoforms in breast cancer progression. We modulated the abundance of specific AKT isoforms in IBH-6 and T47D human breast cancer cell lines and showed that AKT1 promoted cell proliferation, through S6 and cyclin D1 upregulation, but it inhibited cell migration and invasion through ß1-integrin and focal adhesion kinase (FAK) downregulation. In contrast, AKT2 promoted cell migration and invasion through F-actin and vimentin induction. Thus, while overexpression of AKT1 promoted local tumor growth, downregulation of AKT1 or overexpression of AKT2 promoted peritumoral invasion and lung metastasis. Furthermore, we evaluated The Cancer Genome Atlas (TCGA) dataset for invasive breast carcinomas and found that increased AKT2 but not AKT1 mRNA levels correlated with a worse clinical outcome. We conclude that AKT isoforms play specific roles in different steps of breast cancer progression, with AKT1 involved in the local tumor growth and AKT2 involved in the distant tumor dissemination, having AKT2 a poorer prognostic value and consequently being a worthwhile target for therapy.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-akt/genetics , Actins/genetics , Actins/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Disease Progression , Female , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/metabolism , Survival Analysis , Transplantation, Heterologous
9.
Oncotarget ; 6(26): 22081-97, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26098779

ABSTRACT

Improved efficacy of neoadjuvant endocrine-targeting therapies in luminal breast carcinomas could be achieved with optimal use of pathway targeting agents. In a mouse model of ductal breast carcinoma we identify a tumor regressive stromal reaction that is induced by neoadjuvant endocrine therapy. This reparative reaction is characterized by tumor neovascularization accompanied by infiltration of immune cells and carcinoma-associated fibroblasts that stain for phosphorylated ribosomal protein S6 (pS6), downstream the PI3K/Akt/mTOR pathway. While tumor variants with higher PI3K/Akt/mTOR activity respond well to a combination of endocrine and PI3K/Akt/mTOR inhibitors, tumor variants with lower PI3K/Akt/mTOR activity respond more poorly to the combination therapy than to the endocrine therapy alone, associated with inhibition of stromal pS6 and the reparative reaction. In human breast cancer xenografts we confirm that such differential sensitivity to therapy is primarily determined by the level of PI3K/Akt/mTOR in tumor cells. We further show that the clinical response of breast cancer patients undergoing neoadjuvant endocrine therapy is associated with the reparative stromal reaction. We conclude that tumor level and localization of pS6 are associated with therapeutic response in breast cancer and represent biomarkers to distinguish which tumors will benefit from the incorporation of PI3K/Akt/mTOR inhibitors with neoadjuvant endocrine therapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Aged , Aged, 80 and over , Androstadienes/pharmacology , Animals , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Estrogen Receptor Modulators/pharmacology , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Middle Aged , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , Stromal Cells/enzymology , Stromal Cells/pathology , Wortmannin , Xenograft Model Antitumor Assays
10.
Int J Cancer ; 136(11): 2680-92, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25363551

ABSTRACT

There is emerging interest in understanding the role of progesterone receptors (PRs) in breast cancer. The aim of this study was to investigate the proliferative effect of progestins and antiprogestins depending on the relative expression of the A (PRA) and B (PRB) isoforms of PR. In mifepristone (MFP)-resistant murine carcinomas antiprogestin responsiveness was restored by re-expressing PRA using demethylating agents and histone deacetylase inhibitors. Consistently, in two human breast cancer xenograft models, one manipulated to overexpress PRA or PRB (IBH-6 cells), and the other expressing only PRA (T47D-YA) or PRB (T47D-YB), MFP selectively inhibited the growth of PRA-overexpressing tumors and stimulated IBH-6-PRB xenograft growth. Furthermore, in cells with high or equimolar PRA/PRB ratios, which are stimulated to proliferate in vitro by progestins, and are inhibited by MFP, MPA increased the interaction between PR and the coactivator AIB1, and MFP favored the interaction between PR and the corepressor SMRT. In a PRB-dominant context in which MFP stimulates and MPA inhibits cell proliferation, the opposite interactions were observed. Chromatin immunoprecipitation assays in T47D cells in the presence of MPA or MFP confirmed the interactions between PR and the coregulators at the CCND1 and MYC promoters. SMRT downregulation by siRNA abolished the inhibitory effect of MFP on MYC expression and cell proliferation. Our results indicate that antiprogestins are therapeutic tools that selectively inhibit PRA-overexpressing tumors by increasing the SMRT/AIB1 balance at the CCND1 and MYC promoters.


Subject(s)
Cyclin D1/genetics , Mammary Neoplasms, Experimental/drug therapy , Mifepristone/pharmacology , Progestins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Receptors, Progesterone/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Coactivator 3/metabolism , Promoter Regions, Genetic
11.
Carcinogenesis ; 33(3): 509-18, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22180571

ABSTRACT

Using a model of medroxyprogesterone acetate (MPA)-induced mouse mammary tumors that transit through different stages of hormone dependence, we previously reported that the activation of the phosphatidylinositol 3-kinase (PI3K)/AKT (protein kinase B) pathway is critical for the growth of hormone-independent (HI) mammary carcinomas but not for the growth of hormone-dependent (HD) mammary carcinomas. The objective of this work was to explore whether the activation of the PI3K/AKT pathway is responsible for the changes in tumor phenotype and for the transition to autonomous growth. We found that the inhibition of the PI3K/AKT/mTOR (mammalian target of rapamycin) pathway suppresses HI tumor growth. In addition, we were able to induce mammary tumors in mice in the absence of MPA by inoculating HD tumor cells expressing a constitutively active form of AKT1, myristoylated AKT1 (myrAKT1). These tumors were highly differentiated and displayed a ductal phenotype with laminin-1 and cytokeratin 8 expression patterns typical of HI tumors. Furthermore, myrAKT1 increased the tumor growth of IBH-6 and IBH-7 human breast cancer cell lines. In the estrogen-dependent IBH-7 cell line, myrAKT1 induced estrogen-independent growth accompanied by the expression of the adhesion markers focal adhesion kinase and E-cadherin. Finally, we found that cells expressing myrAKT1 exhibited increased phosphorylation of the progesterone receptor at Ser190 and Ser294 and of the estrogen receptor α at Ser118 and Ser167, independently of exogenous MPA or estrogen supply. Our results indicate that the activation of the PI3K/AKT/mTOR pathway promotes tissue architecture remodeling and the activation of steroid receptors.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Animals , Cadherins/biosynthesis , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Female , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Humans , Keratin-8/biosynthesis , Laminin/biosynthesis , Mammary Neoplasms, Experimental/chemically induced , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
12.
PLoS One ; 5(5): e10786, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20520761

ABSTRACT

BACKGROUND: A significant proportion of breast cancer patients face failure of endocrine therapy due to the acquisition of endocrine resistance. We have explored mechanisms involved in such disease progression by using a mouse breast cancer model that is induced by medroxyprogesterone acetate (MPA). These tumors transit through different stages of hormone sensitivity. However, when cells from tumor variants were seeded on plastic, all were stimulated by progestins and inhibited by antiprogestins such as RU486. Furthermore, cells from a RU486-resistant tumor variant recovered antiprogestin sensitivity. HYPOTHESIS: A three-dimensional (3D) culture system, by maintaining differential cellular organization that is typical of each tumor variant, may allow for the maintenance of particular hormone responses and thus be appropriate for the study of the effects of specific inhibitors of signaling pathways associated with disease progression. METHOD: We compared the behavior of tumors growing in vivo and cancer cells ex vivo (in 3D Matrigel). In this system, we evaluated the effects of kinase inhibitors and hormone antagonists on tumor growth. PRINCIPAL FINDINGS: LY294002, a PI3K/AKT pathway inhibitor, decreased both tumor growth in vivo and cell survival in Matrigel in MPA-independent tumors with higher AKT activity. Induction of cell death by anti-hormones such as ICI182780 and ZK230211 was more effective in MPA-dependent tumors with lower AKT activity. Inhibition of MEK with PD98059 did not affect tumor growth in any tested variant. Finally, while Matrigel reproduced differential responsiveness of MPA-dependent and -independent breast cancer cells, it was not sufficient to preserve antiprogestin resistance of RU486-resistant tumors. CONCLUSION: We demonstrated that the PI3K/AKT pathway is relevant for MPA-independent tumor growth. Three-dimensional cultures were useful to test the effects of kinase inhibitors on breast cancer growth and highlight the need for in vivo models to validate experimental tools used for selective therapeutic targeting.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Cell Culture Techniques/methods , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Separation , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Estrogen Receptor alpha/metabolism , Female , Hormones , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Steroid/antagonists & inhibitors , Signal Transduction/drug effects
13.
Breast Cancer Res Treat ; 116(3): 449-60, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18677559

ABSTRACT

To explore mechanisms related to hormone resistance, three resistant variants of the MPA mouse breast cancer tumor model with low levels of progesterone receptor (PR) isoform A (PR-A)/high PR-B expression were developed by prolonged selective pressure with antiprogestins. The resistant phenotype of one tumor line was reversed spontaneously after several consecutive passages in syngeneic BALB/c mice or by 17-beta-estradiol or tamoxifen treatment, and this reversion was significantly associated with an increase in PR-A expression. The responsive parental tumors disclosed low activation of ERK and high activation of AKT; resistant tumors on the other hand, showed the opposite, and this was associated with a higher metastatic potential, that did not revert. This study shows for the first time in vivo a relationship between PR isoform expression and antiprogestin responsiveness, demonstrating that, whereas acquired resistance may be reversed, changes in kinase activation and metastatic potential are unidirectional associated with tumor progression.


Subject(s)
Drug Resistance, Neoplasm , Hormone Antagonists/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mifepristone/pharmacology , Receptors, Progesterone/metabolism , Animals , Blotting, Western , Cell Proliferation , Female , Fluorescent Antibody Technique , Lymphatic Metastasis , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Isoforms , Proto-Oncogene Proteins c-akt/metabolism , Survival Rate
14.
Int J Cancer ; 123(11): 2518-31, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18767044

ABSTRACT

The mechanisms by which mammary carcinomas acquire hormone independence are still unknown. To study the role of cancer-associated fibroblasts (CAF) in the acquisition of hormone-independence we used a hormone-dependent (HD) mouse mammary tumor and its hormone-independent (HI) variant, which grows in vivo without hormone supply. HI tumors express higher levels of FGFR-2 than HD tumors. In spite of their in vivo differences, both tumors have the same hormone requirement in primary cultures. We demonstrated that CAF from HI tumors (CAF-HI) growing in vitro, express higher levels of FGF-2 than HD counterparts (CAF-HD). FGF-2 activated the progesterone receptors (PR) in the tumor cells, thus increasing cell proliferation in both HI and HD tumors. CAF-HI induced a higher proliferative rate on the tumor cells and in PR activation than CAF-HD. The blockage of FGF-2 in the co-cultures or the genetic or pharmacological inhibition of FGFR-2 inhibited PR activation and tumor cell proliferation. Moreover, in vivo, the FGFR inhibitor decreased C4-HI tumor growth, whereas FGF-2 was able to stimulate C4-HD tumor growth as MPA. T47D human breast cancer cells were also stimulated by progestins, FGF-2 or CAF-HI, and this stimulation was abrogated by antiprogestins, suggesting that the murine C4-HI cells respond as the human T47D cells. In summary, this is the first study reporting differences between CAF from HD and HI tumors suggesting that CAF-HI actively participate in driving HI tumor growth.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fibroblast Growth Factor 2/pharmacology , Progesterone/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, Progesterone/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Female , Fibroblasts , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Protein Binding , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction/drug effects
15.
Clin Cancer Res ; 10(1 Pt 2): 402S-9S, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14734499

ABSTRACT

Interactions between luminal epithelial cells and their surrounding microenvironment govern the normal development and function of the mammary gland. Alterations of these interactions can induce abnormal intracellular signaling pathways that affect the development and progression of breast tumors. One critical component of mammary gland development, as well as breast cancer progression, is the expression of estrogen receptors. In a previous study using cultured nonmalignant mammary epithelial cells, we found that the basement membrane molecules, laminin-1 and collagen-IV, were involved in maintenance of estrogen receptor (ER) alpha expression, and that this response could be interfered with by disrupting cell-extracellular matrix adhesion. Here we use phenotypically normal mammary epithelial SCp2 cells to dissect the promoter region of the ERalpha that is involved in the selective response to basement membrane. We also analyze the alteration of this response in SCg6 cells, a malignant cell line that shares a common lineage with the SCp2 cells, to provide insight into the relative overexpression of ERalpha and the unresponsiveness to basement membrane regulation found in those malignant cells. Evidence is presented to show the relevance of the cross-talk between different signaling pathways in the constitution of a functional tissue organization and how this integration may be disrupted in the malignant phenotype.


Subject(s)
Gene Expression Regulation, Developmental , Receptors, Estrogen/metabolism , Animals , Estrogen Receptor alpha , Extracellular Matrix/metabolism , Female , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/therapy , Mice , Mice, Inbred BALB C , Models, Genetic , Phenotype , Promoter Regions, Genetic , Signal Transduction , Time Factors , Transcription, Genetic
16.
J Cell Sci ; 116(Pt 14): 2975-86, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12808020

ABSTRACT

The expression level and functional activity of estrogen receptor alpha is an important determinant of breast physiology and breast cancer treatment. However, it has been difficult to identify the signals that regulate estrogen receptor because cultured mammary epithelial cells generally do not respond to estrogenic signals. Here, we use a combination of two- and three-dimensional culture systems to dissect the extracellular signals that control endogenous estrogen receptor alpha. Its expression was greatly reduced when primary mammary epithelial cells were placed on tissue culture plastic; however, the presence of a reconstituted basement membrane in combination with lactogenic hormones partially prevented this decrease. Estrogen receptor alpha expression in primary mammary fibroblasts was not altered by these culture conditions, indicating that its regulation is cell type specific. Moreover, estrogen receptor-dependent reporter gene expression, as well as estrogen receptor alpha levels, were increased threefold in a functionally normal mammary epithelial cell line when reconstituted basement membrane was added to the medium. This regulatory effect of reconstituted basement membrane was reproduced by two of its components, collagen-IV and laminin-1, and it was blocked by antibodies against alpha2, alpha6 and beta1 integrin subunits. Our results indicate that integrin-mediated response to specific basement membrane components, rather than cell rounding or cell growth arrest induced by reconstituted basement membrane, is critical in the regulation of estrogen receptor alpha expression and function in mammary epithelial cells.


Subject(s)
Collagen Type IV/physiology , Epithelial Cells/metabolism , Laminin/physiology , Mammary Glands, Animal/metabolism , Receptors, Estrogen/biosynthesis , Animals , Basement Membrane/metabolism , Blotting, Western , Caseins/metabolism , Cell Division , Cell Line , Cells, Cultured , Chloramphenicol O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Estrogen Receptor alpha , Fibroblasts/metabolism , Gene Expression Regulation , Integrin alpha2/metabolism , Integrin alpha6/metabolism , Integrin beta1/metabolism , Integrins/metabolism , Ligands , Mice , Microscopy, Fluorescence , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors
17.
Breast Cancer Res ; 5(1): 53-6, 2003.
Article in English | MEDLINE | ID: mdl-12559047

ABSTRACT

The Era of Hope meeting addressed with a multidisciplinary approach the most critical issues in breast carcinogenesis. The issues that we summarize here include: a) the use of rodent models for the study of mammary gland development and breast tumorigenesis; b) the effects of stroma on mammary epithelial differentiation and malignant transformation; c) a further characterization of the interactions between steroid and growth factor receptors; d) the improvement of technologies for early detection of breast tumors and the establishment of their progression; and e) the development of vaccines as potential new therapies against specific tumor markers.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Breast/pathology , Breast Neoplasms/genetics , Cancer Vaccines/therapeutic use , DNA Repair , Female , Humans , Neovascularization, Pathologic/pathology
18.
Fertil Steril ; 78(6): 1278-87, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12477525

ABSTRACT

OBJECTIVE: To evaluate a reciprocal signaling interaction initiated by embryo-derived nitric oxide (NO) to facilitate implantation by increased production of gelatinase A (matrix metalloproteinase 2, MMP2) in uterine stroma. DESIGN: Experimental animal studies. SETTING: Reproductive-physiology research laboratory. ANIMAL(S): Female syngeneic Wistar rats aged 14 weeks. INTERVENTION(S): Vaginal smears to confirm pregnancy. Oviductal ligature to avoid the descent of blastocysts to the uterine lumen. Plasma exudation assays to locate uterine blastocyst implantation sites. Organ cultures treated with NO donors and nitric oxide synthase (NOS) inhibitors. MAIN OUTCOME MEASURE(S): Expression of MMP2 and NO was assessed by Western blot and zymography of tissue extracts and by immunofluorescence of tissue sections. RESULT(S): An increase in MMP2 activity was found in uterine extracts in early pregnant rats and was concentrated at implantation sites. Immunolocalization experiments showed that inducible NOS was expressed on the surface of the implanting blastocyst adjacent to the uterine epithelium at the sites of increased MMP2 expression. In organ culture experiments, NO donors were found to increase, whereas NOS inhibitors were found to decrease MMP2 activity in uterine tissue sections. CONCLUSION(S): Blastocyst-derived NO contributes to the production of uterine-derived MMP2, an essential component of implantation and initiation of placentation.


Subject(s)
Blastocyst/metabolism , Embryo Implantation/physiology , Matrix Metalloproteinase 2/metabolism , Nitric Oxide/physiology , Uterus/enzymology , Animals , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Female , Matrix Metalloproteinase Inhibitors , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Organ Culture Techniques , Pregnancy , Rats , Rats, Wistar , omega-N-Methylarginine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL