Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
PLoS One ; 10(9): e0137920, 2015.
Article En | MEDLINE | ID: mdl-26360292

Multicellular tumor spheroids represent a 3D in vitro model that mimics solid tumor essential properties including assembly and development of extracellular matrix and nutrient, oxygen and proliferation gradients. In the present study, we analyze the impact of 3D spatial organization of HER2-overexpressing breast cancer cells on the response to Trastuzumab. We cultured human mammary adenocarcinoma cell lines as spheroids with the hanging drop method and we observed a gradient of proliferating, quiescent, hypoxic, apoptotic and autophagic cells towards the inner core. This 3D organization decreased Trastuzumab sensitivity of HER2 over-expressing cells compared to monolayer cell cultures. We did not observe apoptosis induced by Trastuzumab but found cell arrest in G0/G1 phase. Moreover, the treatment downregulated the basal apoptosis only found in tumor spheroids, by eliciting protective autophagy. We were able to increase sensitivity to Trastuzumab by autophagy inhibition, thus exposing the interaction between apoptosis and autophagy. We confirmed this result by developing a resistant cell line that was more sensitive to autophagy inhibition than the parental BT474 cells. In summary, the development of Trastuzumab resistance relies on the balance between death and survival mechanisms, characteristic of 3D cell organization. We propose the use of spheroids to further improve the understanding of Trastuzumab antitumor activity and overcome resistance.


Antineoplastic Agents/pharmacology , Autophagy/drug effects , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/genetics , Breast Neoplasms/genetics , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Humans , Phenotype , Receptor, ErbB-2/genetics , Spheroids, Cellular , Trastuzumab/toxicity , Tumor Cells, Cultured
2.
J Immunol ; 194(7): 3452-62, 2015 Apr 01.
Article En | MEDLINE | ID: mdl-25740944

Myeloid-derived suppressor cells (MDSCs) are key regulatory cells that control inflammation and promote tumor-immune escape. To date, no specific immunomodulatory drug has proven efficacy in targeting the expansion and/or function of these cells in different pathophysiologic settings. In this study, we identified a context-dependent effect of the nonsteroidal anti-inflammatory drug indomethacin (IND) on MDSCs, depending on whether they were derived from tumor microenvironments (TME) or from tumor-free microenvironments (TFME). Treatment of mice bearing the LP07 lung adenocarcinoma with IND inhibited the suppressive activity of splenic MDSCs, which restrained tumor growth through mechanisms involving CD8(+) T cells. The same effect was observed when MDSCs were treated with IND and conditioned media from LP07 tumor cells in vitro. However, in the absence of a tumor context, IND enhanced the intrinsic suppressive function of MDSCs and amplified their protumoral activity. In a model of autoimmune neuroinflammation, IND-treated MDSCs differentiated in TFME attenuated inflammation, whereas IND-treated MDSCs differentiated in TME aggravated clinical symptoms and delayed resolution of the disease. Mechanistically, IND reduced arginase activity as well as NO and reactive oxygen species production in MDSCs differentiated in TME but not in TFME. Moreover, expression of the C/EBP-ß transcription factor isoforms correlated with the suppressive activity of IND-treated MDSCs. Our study unveils the dual and context-dependent action of IND, a drug that serves both as an anti-inflammatory and anticancer agent, which differentially affects MDSC activity whether these cells are derived from TME or TFME. These results have broad clinical implication in cancer, chronic inflammation and autoimmunity.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cellular Microenvironment/drug effects , Cellular Microenvironment/immunology , Indomethacin/pharmacology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Autoimmunity/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Immunophenotyping , Mice , Models, Biological , Myeloid Cells/metabolism , Neoplasms/immunology , Neoplasms/pathology , Nitric Oxide/metabolism , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology
3.
Int J Breast Cancer ; 2011: 352182, 2011.
Article En | MEDLINE | ID: mdl-22295219

The epidermal growth factor receptor 2 (HER2) is a tyrosine kinase overexpressed in nearly 20% to 25% of invasive breast cancers. Trastuzumab is a humanized monoclonal antibody that targets HER2. The majority of patients with metastatic breast cancer initially respond to trastuzumab, however, within 1 year of treatment disease progresses. Several molecular mechanisms have been described as contributing to the development of trastuzumab resistance. They could be grouped as impaired access of trastuzumab to HER2, upregulation of HER2 downstream signaling pathways, signaling of alternative pathways, and impaired immune antitumor mechanisms. However, since many of them have overlapping effects, it would be of great clinical impact to identify the principal signaling pathways involved in drug resistance. Significant efforts are being applied to find other therapeutic modalities besides trastuzumab treatment to be used alone or in combination with current modalities.

4.
Nitric Oxide ; 23(3): 234-41, 2010 Nov 01.
Article En | MEDLINE | ID: mdl-20633694

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.


Cobalt/pharmacology , Hypoxia , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Structure-Activity Relationship , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis
5.
Breast Cancer Res ; 7(3): R345-52, 2005.
Article En | MEDLINE | ID: mdl-15987429

INTRODUCTION: The role of macrophages in tumor progression has generated contradictory evidence. We had previously demonstrated the ability of peritoneal macrophages from LMM3 murine mammary adenocarcinoma-bearing mice (TMps) to increase the angiogenicity of LMM3 tumor cells, mainly through polyamine synthesis. Here we investigate the ability of the parasympathetic nervous system to modulate angiogenesis induced by TMps through the activation of the muscarinic acetylcholine receptor (mAchR). METHODS: Peritoneal macrophages from female BALB/c mice bearing a 7-day LMM3 tumor were inoculated intradermally (3 x 10(5) cells per site) into syngeneic mice. Before inoculation, TMps were stimulated with the muscarinic agonist carbachol in the absence or presence of different muscarinic antagonists or enzyme inhibitors. Angiogenesis was evaluated by counting vessels per square millimeter of skin. The expression of mAchR, arginase and cyclo-oxygenase (COX) isoforms was analyzed by Western blotting. Arginase and COX activities were evaluated by urea and prostaglandin E2 (PGE2) production, respectively. RESULTS: TMps, which stimulate neovascularization, express functional mAchR, because carbachol-treated TMps potently increased new blood vessels formation. This response was completely blocked by preincubating TMps with pirenzepine and 4-diphenylacetoxy-N-methylpiperidine (4-DAMP), M1 and M3 receptor antagonists, and partly by the M2 receptor antagonist methoctramine. M1 receptor activation by carbachol in TMps triggers neovascularization through arginase products because Nomega-hydroxy-L-arginine reversed the agonist action. Preincubation of TMps with methoctramine partly prevented carbachol-stimulated urea formation. In addition, COX-derived liberation of PGE2 is responsible for the promotion of TMps angiogenic activity by M3 receptor. We also detected a higher expression of vascular endothelial growth factor (VEGF) in TMps than in macrophages from normal mice. Carbachol significantly increased VEGF expression in TMps, and this effect was totally reversed by methoctramine and pirenzepine. Arginase and COX inhibitors partly decreased VEGF derived from TMps. CONCLUSION: TMps themselves induce a potent angiogenic response that is augmented by carbachol action. mAchR activation triggers arginine metabolism, PGE2 synthesis and VEGF production, promoting neovascularization.


Adenocarcinoma/physiopathology , Mammary Neoplasms, Animal/physiopathology , Neovascularization, Pathologic , Receptors, Muscarinic/physiology , Animals , Arginase/biosynthesis , Arginase/metabolism , Blotting, Western , Female , Gene Expression Profiling , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Isoforms , Receptors, Muscarinic/biosynthesis
6.
Int J Mol Med ; 9(6): 651-7, 2002 Jun.
Article En | MEDLINE | ID: mdl-12011984

Investigations on the influence of the parasympathetic nervous system via muscarinic signaling in tumor progression have produced contradictory evidence. We investigated the expression of muscarinic acetylcholine receptors (mAchR) and their intracellular transduction pathways, in two murine mammary adenocarcinoma cell lines, LM3 and LM2 in comparison with the normal murine mammary epithelial cell line: NMuMG. Saturation binding assays with the tritiated muscarinic antagonist quinuclidinyl benzilate ([3H]-QNB) indicate that LM3 cells express higher amounts of mAchR than LM2 cells. Muscarinic receptor activation with carbachol (CARB) enhanced basal production of citrulline to a greater extent in LM3 cells than in LM2 cells. The nitric oxide synthase (NOS) inhibitor, NGmono-methyl-L-arginine (L-NMMA), blunted this effect only in LM3 cells while in LM2 cells the action of CARB was blocked by Nomega hydroxy-L-arginine (L-OH-Arg), which is known to inhibit the arginase pathway. Atropine blocks the action of CARB in both cell lines. Additionally, mAchR activation stimulates prostaglandin E2 (PGE2) synthesis only in LM2 cells. NMuMG cells show detectable basal amounts of nitric oxide and PGE2, but they did not respond to CARB. Binding experiments confirm the absence of mAchR in these cells. The findings indicate that mAchR expression in tumor cells, and its control on arginine metabolism, via NOS/arginase, and on PGE2 synthesis by COX activation, could be a switch on mechanism that might lead mammary cells from normal to malignant phenotype. Moreover, mAchR coupling to distinct effectors might be associated with differences in aggressiveness of tumor cells.


Adenocarcinoma/enzymology , Arginase/biosynthesis , Mammary Neoplasms, Animal/metabolism , Nitric Oxide Synthase/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Receptors, Muscarinic/metabolism , Animals , Blotting, Western , Carbachol/pharmacology , Cell Division , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase/chemistry , Protein Binding , Protein Isoforms , Receptors, Muscarinic/biosynthesis , Tumor Cells, Cultured
7.
Int J Oncol ; 20(1): 59-67, 2002 Jan.
Article En | MEDLINE | ID: mdl-11743643

Regional lymph nodes are important in the generation of tumor-directed immune responses. The relationship between nitric oxide synthase (NOS) expression and the biological behavior of tumor-draining lymph node (TDLNs) cells in vivo was determined using a spontaneously arising BALB/c mammary adenocarcinoma S13. We first demonstrated a reduction of tumor size and tumor-induced angiogenesis by blocking NOS activity in vivo. TDLNs harvested from tumor-bearing mice (TBM) on day 16 after tumor implant, showed enhanced NOS activity and NOS expression compared to control nodes. Identification of the NOS isoforms present in TDLNs resulted in expression of neuronal NOS (nNOS), endothelial NOS (eNOS) and absence of inducible NOS (iNOS). TDLN cells admixed with tumor cells and inoculated into normal mice (Winn assay) induced a reduction of tumor growth although, when inoculated alone, were able to induce the formation of new blood vessels (angiogenesis). Our data indicate that the in vivo antitumor activity of TDLN cells is modulated by a balance between angiogenesis and antitumor effectors. In our model, when trafficking of leukocytes is obviated, the control of tumor growth by TDLN cells can be explained in part by an antitumor activity great enough to exceed the angiogenic component elicited by the same cells, leading to a reduction of tumor size.


Adenocarcinoma/prevention & control , Lymph Nodes/enzymology , Mammary Neoplasms, Experimental/prevention & control , Nitric Oxide Synthase/metabolism , Nitric Oxide/physiology , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Animals , Enzyme Inhibitors/pharmacology , Immunoblotting , Lymph Nodes/drug effects , Lymphatic Metastasis , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , NG-Nitroarginine Methyl Ester/pharmacology , Neovascularization, Pathologic/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III
8.
Medicina [B.Aires] ; 49(3): 265-70, mayo-jun. 1989. Tab
Article Es | BINACIS | ID: bin-28104

Estudiamos la regulación del crecimiento tumoral y metastásico tanto por el propio tumor como por el sistema inmune, en un modelo murino. Sobrenadantes de cultivo de esplenocitos de portadores de tumor exacerban el crecimiento del propio tumor. Esta actividad desaparece luego de la cirugía tumoral pero con una cinética diferente dependendo de si el tumor es precoz o avanzado al tiempo de la cirugía. Las poblaciones esplénicas involucradas en la exacerbación varían durante el crecimiento del tumor mientras que las responsables de inducción de angiogénesis son siempre de naturaleza T. Con respecto de la autorregulación tumoral, encontramos que diferentes formas de antígenos tumorales (extractos tumorales, sobrenadantes de cultivo de células tumorales, células tumorales formolizadas) aumentan la diseminación metastásica, pero esta actividad está mediada por el sistema inmune del huésped. Por el contrario, sobrenadantes de cultivo de células tumorales no inducen exacerbación del tumor primario. Las células tumorales fueron tratadas con un modificador de membrana con la expectativa de alterar la respuesta inmune antitumoral (AU)


Mice , Animals , Adenocarcinoma/immunology , Mammary Neoplasms, Experimental/immunology , Spleen/cytology , Antigens, Neoplasm/immunology , Lymphocytes/physiology , Adenocarcinoma/secondary , Adenocarcinoma/pathology , Mammary Neoplasms, Experimental/secondary , Mammary Neoplasms, Experimental/pathology
9.
Medicina (B.Aires) ; 49(3): 265-70, mayo-jun. 1989. tab
Article Es | LILACS | ID: lil-86681

Estudiamos la regulación del crecimiento tumoral y metastásico tanto por el propio tumor como por el sistema inmune, en un modelo murino. Sobrenadantes de cultivo de esplenocitos de portadores de tumor exacerban el crecimiento del propio tumor. Esta actividad desaparece luego de la cirugía tumoral pero con una cinética diferente dependendo de si el tumor es precoz o avanzado al tiempo de la cirugía. Las poblaciones esplénicas involucradas en la exacerbación varían durante el crecimiento del tumor mientras que las responsables de inducción de angiogénesis son siempre de naturaleza T. Con respecto de la autorregulación tumoral, encontramos que diferentes formas de antígenos tumorales (extractos tumorales, sobrenadantes de cultivo de células tumorales, células tumorales formolizadas) aumentan la diseminación metastásica, pero esta actividad está mediada por el sistema inmune del huésped. Por el contrario, sobrenadantes de cultivo de células tumorales no inducen exacerbación del tumor primario. Las células tumorales fueron tratadas con un modificador de membrana con la expectativa de alterar la respuesta inmune antitumoral


Mice , Animals , Adenocarcinoma/immunology , Mammary Neoplasms, Experimental/immunology , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Antigens, Neoplasm/immunology , Lymphocytes/physiology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Spleen/cytology
...