Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
2.
Invest New Drugs ; 40(1): 30-41, 2022 02.
Article in English | MEDLINE | ID: mdl-34478029

ABSTRACT

Breast cancer is the leading cause of cancer death among women worldwide. For this reason, the development of new therapies is still essential. In this work we have analyzed the antitumor potential of levoglucosenone, a chiral building block derived from the pyrolysis of cellulose-containing materials such as soybean hulls, and three structurally related analogues. Employing human and murine mammary cancer models, we have evaluated the effect of our compounds on cell viability through MTS assay, apoptosis induction by acridine orange/ethidium bromide staining and/or flow cytometry and the loss of mitochondrial potential by tetramethylrhodamine methyl ester staining. Autophagy and senescence induction were also evaluated by Western blot and ß-galactosidase activity respectively. Secreted metalloproteases activity was determined by quantitative zymography. Migratory capacity was assessed by wound healing assays while invasive potential was analyzed using Matrigel-coated transwell chambers. In vivo studies were also performed to evaluate subcutaneous tumor growth and experimental lung colonization. All compounds impaired in vitro proliferation with IC50 values in a range of low micromolar. Apoptosis was identified as the main mechanism responsible for the reduction of monolayer cell content induced by the compounds without detecting modulations of autophagy or senescence processes. Two of the four compounds (levoglucosenone and its brominated variant) were able to modulate in vitro events associated with tumor progression, such as migratory potential, invasiveness, and proteases secretion. Furthermore, tumor volume and metastatic spread were significantly reduced in vivo after the treatment these two compounds. Here, we could obtain from soybean hulls, a material with almost no commercial value, a variety of chemical compounds useful for breast cancer treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Glucose/analogs & derivatives , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellulose/chemistry , Dose-Response Relationship, Drug , Glucose/chemistry , Glucose/pharmacology , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Tumor Burden/drug effects
3.
J Cancer Res Clin Oncol ; 146(12): 3241-3253, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32865619

ABSTRACT

PURPOSE: Retinoids have proved to be effective for hematologic malignancies treatment but till nowadays, their use as single agent for the solid tumor's management is still controversial. All-trans retinoic acid (ATRA), the main active metabolite of vitamin A, exerts non-genomic interactions with different members of the protein kinase C (PKC) family, recognized modulators of different tumor progression pathways. To determine whether a group of patients could become benefited employing a retinoid therapy, in this study we have evaluated whether PKCα expression (a poor prognosis marker in breast cancer) could sensitizes mammary cells to ATRA treatment. METHODS: PKCα overexpression was achieved by stable transfection and confirmed by western blot. Transfected PKC functionality was determined by nuclear translocation-induction and confocal microscopy. In vitro proliferation was evaluated by cell counting and cell cycle distribution was analyzed by flow cytometry. In vivo studies were performed to evaluate orthotopic tumor growth and experimental lung colonization. Retinoic acid response elements (RARE) and AP1 sites-dependent activity was studied by gene reporter assays and retinoic acid receptors (RARs) were measured by RT-qPCR. RESULTS: Our findings suggest that high PKCα levels improve the differentiation response to ATRA in a RAR signaling-dependent manner. Moreover, RARß expression appears to be critical to induce ATRA sensitization, throughout AP1 trans-repression. CONCLUSION: Here we propose that retinoids could lead a highly personalized anticancer treatment, bringing benefits to patients with aggressive breast tumors resulting from high PKCα expression but, an adequate expression of the RARß receptor is required to ensure the effect on this process.


Subject(s)
Breast Neoplasms/drug therapy , Protein Kinase C-alpha/genetics , Receptors, Retinoic Acid/genetics , Tretinoin/pharmacology , Animals , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , MCF-7 Cells , Mice , Retinoids/pharmacology , Signal Transduction/drug effects , Vitamin A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...