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.
Cancer Res ; 72(22): 5856-66, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23010075

ABSTRACT

The mammary ducts of humans and mice are comprised of two main mammary epithelial cell (MEC) subtypes: a surrounding layer of basal MECs and an inner layer of luminal MECs. Breast cancer subtypes show divergent clinical behavior that may reflect properties inherent in their MEC compartment of origin. How the response to a cancer-initiating genetic event is shaped by MEC subtype remains largely unexplored. Using the mouse mammary gland, we designed organotypic three-dimensional culture models that permit challenge of discrete MEC compartments with the same oncogenic insult. Mammary organoids were prepared from mice engineered for compartment-restricted coexpression of oncogenic H-RAS(G12V) together with a nuclear fluorescent reporter. Monitoring of H-RAS(G12V)-expressing MECs during extended live cell imaging permitted visualization of Ras-driven phenotypes via video microscopy. Challenging either basal or luminal MECs with H-RAS(G12V) drove MEC proliferation and survival, culminating in aberrant organoid overgrowth. In each compartment, Ras activation triggered modes of collective MEC migration and invasion that contrasted with physiologic modes used during growth factor-initiated branching morphogenesis. Although basal and luminal Ras activation produced similar overgrowth phenotypes, inhibitor studies revealed divergent use of Ras effector pathways. Blocking either the phosphoinositide 3-kinase or the mammalian target of rapamycin pathway completely suppressed Ras-driven invasion and overgrowth of basal MECs, but only modestly attenuated Ras-driven phenotypes in luminal MECs. We show that MEC subtype defines signaling pathway dependencies downstream of Ras. Thus, cells-of-origin may critically determine the drug sensitivity profiles of mammary neoplasia.


Subject(s)
Mammary Glands, Animal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism , Animals , Benzamides/pharmacology , Cell Growth Processes/physiology , Chromones/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Immunohistochemistry , Mammary Glands, Animal/cytology , Mammary Glands, Animal/drug effects , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transgenes , ras Proteins/genetics
2.
Muscle Nerve ; 46(3): 407-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22907232

ABSTRACT

INTRODUCTION AND METHODS: This study compared changes in myokine and myogenic genes following resistance exercise (3 sets of 12 repetitions of maximal unilateral knee extension) in 20 elderly men (67.8 ± 1.0 years) and 15 elderly women (67.2 ± 1.5 years). RESULTS: Monocyte chemotactic protein (MCP)-1, macrophage inhibitory protein (MIP)-1ß, interleukin (IL)-6 and MyoD mRNA increased significantly (P < 0.05), whereas myogenin and myostatin mRNA decreased significantly after exercise in both groups. Macrophage-1 (Mac-1) and MCP-3 mRNA did not change significantly after exercise in either group. MIP-1ß, Mac-1 and myostatin mRNA were significantly higher before and after exercise in men compared with women. In contrast, MCP-3 and myogenin mRNA were significantly higher before and after exercise in the women compared with the men. CONCLUSIONS: In elderly individuals, gender influences the mRNA expression of certain myokines and growth factors, both at rest and after resistance exercise. These differences may influence muscle regeneration following muscle injury.


Subject(s)
Exercise/physiology , Inflammation/metabolism , Muscle, Skeletal/metabolism , Resistance Training , Aged , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL4/genetics , Chemokine CCL4/metabolism , Chemokine CCL7/genetics , Chemokine CCL7/metabolism , Female , Gene Expression , Humans , Inflammation/physiopathology , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , Male , Middle Aged , Muscle Development , Muscle, Skeletal/physiopathology , Myogenin/genetics , Myogenin/metabolism , Sex Factors
3.
J Clin Invest ; 118(1): 51-63, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18060046

ABSTRACT

Breast cancers frequently progress or relapse during targeted therapy, but the molecular mechanisms that enable escape remain poorly understood. We elucidated genetic determinants underlying tumor escape in a transgenic mouse model of Wnt pathway-driven breast cancer, wherein targeted therapy is simulated by abrogating doxycycline-dependent Wnt1 transgene expression within established tumors. In mice with intact tumor suppressor pathways, tumors typically circumvented doxycycline withdrawal by reactivating Wnt signaling, either via aberrant (doxycycline-independent) Wnt1 transgene expression or via acquired somatic mutations in the gene encoding beta-catenin. Germline introduction of mutant tumor suppressor alleles into the model altered the timing and mode of tumor escape. Relapses occurring in the context of null Ink4a/Arf alleles (disrupting both the p16 Ink4a and p19 Arf tumor suppressors) arose quickly and rarely reactivated the Wnt pathway. In addition, Ink4a/Arf-deficient relapses resembled p53-deficient relapses in that both displayed morphologic and molecular hallmarks of an epithelial-to-mesenchymal transition (EMT). Notably, Ink4a/Arf deficiency promoted relapse in the absence of gross genomic instability. Moreover, Ink4a/Arf-encoded proteins differed in their capacity to suppress oncogene independence. Isolated p19 Arf deficiency mirrored p53 deficiency in that both promoted rapid, EMT-associated mammary tumor escape, whereas isolated p16 Ink4a deficiency failed to accelerate relapse. Thus, p19 Arf/p53 pathway lesions may promote mammary cancer relapse even when inhibition of a targeted oncogenic signaling pathway remains in force.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Mammary Neoplasms, Experimental/metabolism , Tumor Escape/genetics , Tumor Suppressor Protein p53/metabolism , Wnt1 Protein/metabolism , Alleles , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Female , Genomic Instability/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Mutation , Recurrence , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Wnt1 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...