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
1.
PLoS One ; 14(9): e0220973, 2019.
Article in English | MEDLINE | ID: mdl-31536495

ABSTRACT

In breast cancer, tumor hypoxia has been linked to poor prognosis and increased metastasis. Hypoxia activates transcriptional programs in cancer cells that lead to increased motility and invasion, as well as various metabolic changes. One of these metabolic changes, an increase in glycogen metabolism, has been further associated with protection from reactive oxygen species damage that may lead to premature senescence. Here we report that breast cancer cells significantly increase glycogen stores in response to hypoxia. We found that knockdown of the brain isoform of an enzyme that catalyzes glycogen breakdown, glycogen phosphorylase B (PYGB), but not the liver isoform, PYGL, inhibited glycogen utilization in estrogen receptor negative and positive breast cancer cells; whereas both independently inhibited glycogen utilization in the normal-like breast epithelial cell line MCF-10A. Functionally, PYGB knockdown and the resulting inhibition of glycogen utilization resulted in significantly decreased wound-healing capability in MCF-7 cells and a decrease in invasive potential of MDA-MB-231 cells. Thus, we identify PYGB as a novel metabolic target with potential applications in the management and/or prevention of metastasis in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Glycogen/metabolism , Hypoxia/metabolism , Phenotype , Phosphorylase b/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Metabolic Networks and Pathways , Neoplasm Metastasis , Neoplasm Staging , Phosphorylase b/genetics , Protein Isoforms , RNA Interference , RNA, Small Interfering/genetics
2.
Lab Chip ; 19(7): 1162-1173, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30810557

ABSTRACT

Brain metastases are the most lethal complication of advanced cancer; therefore, it is critical to identify when a tumor has the potential to metastasize to the brain. There are currently no interventions that shed light on the potential of primary tumors to metastasize to the brain. We constructed and tested a platform to quantitatively profile the dynamic phenotypes of cancer cells from aggressive triple negative breast cancer cell lines and patient derived xenografts (PDXs), generated from a primary tumor and brain metastases from tumors of diverse organs of origin. Combining an advanced live cell imaging algorithm and artificial intelligence, we profile cancer cell extravasation within a microfluidic blood-brain niche (µBBN) chip, to detect the minute differences between cells with brain metastatic potential and those without with a PPV of 0.91 in the context of this study. The results show remarkably sharp and reproducible distinction between cells that do and those which do not metastasize inside of the device.


Subject(s)
Artificial Intelligence , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cell Separation/methods , Cell Line, Tumor , Humans , Phenotype
3.
Sci Rep ; 6: 39190, 2016 12 19.
Article in English | MEDLINE | ID: mdl-27991524

ABSTRACT

Inflammatory breast cancer (IBC) is the most lethal form of breast cancer. All IBC patients have lymph node involvement and one-third of patients already have distant metastasis at diagnosis. This propensity for metastasis is a hallmark of IBC distinguishing it from less lethal non-inflammatory breast cancers (nIBC). Genetic profiling studies have been conducted to differentiate IBC from nIBC, but no IBC cancer-cell-specific gene signature has been identified. We hypothesized that a tumor-extrinsic factor, notably tumor-associated macrophages, promotes and contributes to IBC's extreme metastatic phenotype. To this end, we studied the effect of macrophage-conditioned media (MCM) on IBC. We show that two IBC cell lines are hyper-responsive to MCM as compared to normal-like breast and aggressive nIBC cell lines. We further interrogated IBC's hyper-responsiveness to MCM using a microfluidic migration device, which permits individual cell migration path tracing. We found the MCM "primes" the IBC cells' cellular machinery to become extremely migratory in response to a chemoattractant. We determined that interleukins -6, -8, and -10 within the MCM are sufficient to stimulate this enhanced IBC migration effect, and that the known metastatic oncogene, RhoC GTPase, is necessary for the enhanced migration response.


Subject(s)
Inflammatory Breast Neoplasms/pathology , Macrophages/metabolism , rhoC GTP-Binding Protein/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Chemotactic Factors/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Female , Humans , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/metabolism , Interleukin-10/analysis , Interleukin-10/metabolism , Interleukin-10/pharmacology , Interleukin-6/analysis , Interleukin-6/metabolism , Interleukin-6/pharmacology , Interleukin-8/analysis , Interleukin-8/metabolism , Interleukin-8/pharmacology , Macrophages/cytology , Microfluidics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...