Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Dis Model Mech ; 17(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38506114

ABSTRACT

Breast cancer stands as the most prevalent malignancy afflicting women. Despite significant advancements in its diagnosis and treatment, breast cancer metastasis continues to be a leading cause of mortality among women. To metastasize, cancer cells face numerous challenges: breaking away from the primary tumor, surviving in the circulation, establishing in a distant location, evading immune detection and, finally, thriving to initiate a new tumor. Each of these sequential steps requires cancer cells to adapt to a myriad of stressors and develop survival mechanisms. In addition, most patients with breast cancer undergo surgical removal of their primary tumor and have various therapeutic interventions designed to eradicate cancer cells. Despite this plethora of attacks and stresses, certain cancer cells not only manage to persist but also proliferate robustly, giving rise to substantial tumors that frequently culminate in the patient's demise. To enhance patient outcomes, there is an imperative need for a deeper understanding of the molecular and cellular mechanisms that empower cancer cells to not only survive but also expand. Herein, we delve into the intrinsic stresses that cancer cells encounter throughout the metastatic journey and the additional stresses induced by therapeutic interventions. We focus on elucidating the remarkable strategies adopted by cancer cells, such as cell-cell clustering and intricate cell-cell communication mechanisms, to ensure their survival.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Cell Communication
2.
Oncogene ; 42(10): 737-747, 2023 03.
Article in English | MEDLINE | ID: mdl-36604566

ABSTRACT

Inter-patient and intra-tumoral heterogeneity complicate the identification of predictive biomarkers and effective treatments for basal triple negative breast cancer (b-TNBC). Invasion is the initiating event in metastasis and can occur by both collective and single-cell mechanisms. We cultured primary organoids from a b-TNBC genetically engineered mouse model in 3D collagen gels to characterize their invasive behavior. We observed that organoids from the same tumor presented different phenotypes that we classified as non-invasive, collective and disseminative. To identify molecular regulators driving these invasive phenotypes, we developed a workflow to isolate individual organoids from the collagen gels based on invasive morphology and perform RNA sequencing. We next tested the requirement of differentially regulated genes for invasion using shRNA knock-down. Strikingly, KRAS was required for both collective and disseminative phenotypes. We then performed a drug screen targeting signaling nodes upstream and downstream of KRAS. We found that inhibition of EGFR, MAPK/ERK, or PI3K/AKT signaling reduced invasion. Of these, ERK inhibition was striking for its ability to potently inhibit collective invasion and dissemination. We conclude that different cancer cells in the same b-TNBC tumor can express different metastatic molecular programs and identified KRAS and ERK as essential regulators of collective and single cell dissemination.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Animals , Mice , Triple Negative Breast Neoplasms/pathology , Proto-Oncogene Proteins p21(ras) , Cell Line, Tumor , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Cell Movement/genetics
3.
Sci Transl Med ; 14(656): eabn7571, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35921474

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype associated with early metastatic recurrence and worse patient outcomes. TNBC tumors express molecular markers of the epithelial-mesenchymal transition (EMT), but its requirement during spontaneous TNBC metastasis in vivo remains incompletely understood. We demonstrated that spontaneous TNBC tumors from a genetically engineered mouse model (GEMM), multiple patient-derived xenografts, and archival patient samples exhibited large populations in vivo of hybrid E/M cells that lead invasion ex vivo while expressing both epithelial and mesenchymal characteristics. The mesenchymal marker vimentin promoted invasion and repressed metastatic outgrowth. We next tested the requirement for five EMT transcription factors and observed distinct patterns of utilization during invasion and colony formation. These differences suggested a sequential activation of multiple EMT molecular programs during the metastatic cascade. Consistent with this model, our longitudinal single-cell RNA analysis detected three different EMT-related molecular patterns. We observed cancer cells progressing from epithelial to hybrid E/M and strongly mesenchymal patterns during invasion and from epithelial to a hybrid E/M pattern during colony formation. We next investigated the relative epithelial versus mesenchymal state of cancer cells in both GEMM and patient metastases. In both contexts, we observed heterogeneity between and within metastases in the same individual. We observed a complex spectrum of epithelial, hybrid E/M, and mesenchymal cell states within metastases, suggesting that there are multiple successful molecular strategies for distant organ colonization. Together, our results demonstrate an important and complex role for EMT programs during TNBC metastasis.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Humans , Mice , Neoplasm Metastasis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Vimentin
4.
Cell Metab ; 33(7): 1342-1357.e10, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34102109

ABSTRACT

Mechanical signals from the tumor microenvironment modulate cell mechanics and influence cell metabolism to promote cancer aggressiveness. Cells withstand external forces by adjusting the stiffness of their cytoskeleton. Microtubules (MTs) act as compression-bearing elements. Yet how cancer cells regulate MT dynamic in response to the locally constrained environment has remained unclear. Using breast cancer as a model of a disease in which mechanical signaling promotes disease progression, we show that matrix stiffening rewires glutamine metabolism to promote MT glutamylation and force MT stabilization, thereby promoting cell invasion. Pharmacologic inhibition of glutamine metabolism decreased MT glutamylation and affected their mechanical stabilization. Similarly, decreased MT glutamylation by overexpressing tubulin mutants lacking glutamylation site(s) decreased MT stability, thereby hampering cancer aggressiveness in vitro and in vivo. Together, our results decipher part of the enigmatic tubulin code that coordinates the fine-tunable properties of MT and link cell metabolism to MT dynamics and cancer aggressiveness.


Subject(s)
Glutamic Acid/metabolism , Mechanotransduction, Cellular/physiology , Microtubules/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cells, Cultured , Energy Metabolism/physiology , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Protein Processing, Post-Translational , Tubulin/metabolism , Tumor Microenvironment/physiology
5.
Nat Protoc ; 15(8): 2413-2442, 2020 08.
Article in English | MEDLINE | ID: mdl-32690957

ABSTRACT

Cancer invasion and metastasis are challenging to study in vivo since they occur deep inside the body over extended time periods. Organotypic 3D culture of fresh tumor tissue enables convenient real-time imaging, genetic and microenvironmental manipulation and molecular analysis. Here, we provide detailed protocols to isolate and culture heterogenous organoids from murine and human primary and metastatic site tumors. The time required to isolate organoids can vary based on the tissue and organ type but typically takes <7 h. We describe a suite of assays that model specific aspects of metastasis, including proliferation, survival, invasion, dissemination and colony formation. We also specify comprehensive protocols for downstream applications of organotypic cultures that will allow users to (i) test the role of specific genes in regulating various cellular processes, (ii) distinguish the contributions of several microenvironmental factors and (iii) test the effects of novel therapeutics.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Tissue Culture Techniques/methods , Animals , Humans , Mice , Neoplasm Metastasis
6.
Cell Metab ; 29(1): 124-140.e10, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30293773

ABSTRACT

Dysregulation of extracellular matrix (ECM) deposition and cellular metabolism promotes tumor aggressiveness by sustaining the activity of key growth, invasion, and survival pathways. Yet mechanisms by which biophysical properties of ECM relate to metabolic processes and tumor progression remain undefined. In both cancer cells and carcinoma-associated fibroblasts (CAFs), we found that ECM stiffening mechanoactivates glycolysis and glutamine metabolism and thus coordinates non-essential amino acid flux within the tumor niche. Specifically, we demonstrate a metabolic crosstalk between CAF and cancer cells in which CAF-derived aspartate sustains cancer cell proliferation, while cancer cell-derived glutamate balances the redox state of CAFs to promote ECM remodeling. Collectively, our findings link mechanical stimuli to dysregulated tumor metabolism and thereby highlight a new metabolic network within tumors in which diverse fuel sources are used to promote growth and aggressiveness. Furthermore, this study identifies potential metabolic drug targets for therapeutic development in cancer.


Subject(s)
Aspartic Acid/metabolism , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Carcinoma/metabolism , Glutamic Acid/metabolism , Head and Neck Neoplasms/metabolism , Lung Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Cell Line , Extracellular Matrix , Female , Humans , Mice , Mice, Inbred BALB C , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
7.
Cancer Res ; 78(18): 5229-5242, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30026329

ABSTRACT

In squamous cell carcinoma (SCC), tissue invasion by collectively invading cells requires physical forces applied by tumor cells on their surrounding extracellular matrix (ECM). Cancer-related ECM is composed of thick collagen bundles organized by carcinoma-associated fibroblasts (CAF) within the tumor stroma. Here, we show that SCC cell collective invasion is driven by the matrix-dependent mechano-sensitization of EGF signaling in cancer cells. Calcium (Ca2+) was a potent intracellular second messenger that drove actomyosin contractility. Tumor-derived matrix stiffness and EGFR signaling triggered increased intracellular Ca2+ through CaV1.1 expression in SCC cells. Blocking L-type calcium channel expression or activity using Ca2+ channel blockers verapamil and diltiazem reduced SCC cell collective invasion both in vitro and in vivo These results identify verapamil and diltiazem, two drugs long used in medical care, as novel therapeutic strategies to block the tumor-promoting activity of the tumor niche.Significance: This work demonstrates that calcium channels blockers verapamil and diltiazem inhibit mechano-sensitization of EGF-dependent cancer cell collective invasion, introducing potential clinical strategies against stromal-dependent collective invasion.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/18/5229/F1.large.jpg Cancer Res; 78(18); 5229-42. ©2018 AACR.


Subject(s)
Calcium Signaling , Carcinoma, Squamous Cell/pathology , Extracellular Matrix/metabolism , Head and Neck Neoplasms/pathology , Actomyosin/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Calcium Channels, L-Type , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement , Collagen/metabolism , Diltiazem/pharmacology , ErbB Receptors/metabolism , Fibroblasts/metabolism , Head and Neck Neoplasms/metabolism , Humans , Neoplasm Invasiveness , Spheroids, Cellular , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...