Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Cancer Res ; 82(17): 3045-3057, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35792658

ABSTRACT

Many advanced therapeutics possess cytostatic properties that suppress cancer cell growth without directly inducing death. Treatment-induced cytostatic cancer cells can persist and constitute a reservoir from which recurrent growth and resistant clones can develop. Current management approaches primarily comprise maintenance and monitoring because strategies for targeting nonproliferating cancer cells have been elusive. Here, we used targeted therapy paradigms and engineered cytostatic states to explore therapeutic opportunities for depleting treatment-mediated cytostatic cancer cells. Sustained oncogenic AKT signaling was common, while nonessential, in treatment-mediated cytostatic cancer cells harboring PI3K-pathway mutations, which are associated with cancer recurrence. Engineering oncogenic signals in quiescent mammary organotypic models showed that sustained, aberrant activation of AKT sensitized cytostatic epithelial cells to proteasome inhibition. Mechanistically, sustained AKT signaling altered cytostatic state homeostasis and promoted an oxidative and proteotoxic environment, which imposed an increased proteasome dependency for maintaining cell viability. Under cytostatic conditions, inhibition of the proteasome selectively induced apoptosis in the population with aberrant AKT activation compared with normal cells. Therapeutically exploiting this AKT-driven proteasome vulnerability was effective in depleting treatment-mediated cytostatic cancer cells independent of breast cancer subtype, epithelial origin, and cytostatic agent. Moreover, transient targeting during cytostatic treatment conditions was sufficient to reduce recurrent tumor growth in spheroid and mouse models. This work identified an AKT-driven proteasome-vulnerability that enables depletion of persistent cytostatic cancer cells harboring PTEN-PI3K pathway mutations, revealing a viable strategy for targeting nonproliferating persistent cancer cell populations before drug resistance emerges. SIGNIFICANCE: This study finds that sustained oncogenic signaling in therapy-induced cytostatic cancer cells confers targetable vulnerabilities to deplete persistent cancer cell populations and reduce cancer recurrence.


Subject(s)
Cytostatic Agents , Phosphatidylinositol 3-Kinases , Animals , Apoptosis , Carcinogenesis , Cell Line, Tumor , Cytostatic Agents/pharmacology , Mice , Neoplasm Recurrence, Local , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-akt/metabolism
3.
Methods Mol Biol ; 2429: 405-416, 2022.
Article in English | MEDLINE | ID: mdl-35507177

ABSTRACT

Three-dimensional (3D) cell cultures based on reconstituted basement membrane materials recapitulate features of extracellular matrix (ECM) and tissue stiffness in vivo and provide a physiologically relevant platform to study complex cellular processes, such as stem cell differentiation and tissue morphogenesis, that are otherwise difficult in animal models. The form and composition of 3D matrices in culture can interfere with and pose challenges for different experimental setups and assays, which necessitate alterations to facilitate analysis. Here, we provide a unified protocol for 3D cell cultures with modular workflows that streamline procedures for compatibility with common molecular and cellular assays such as live-cell imaging, immunofluorescence , qPCR, RNAseq, western blotting, and quantitative mass spectrometry.


Subject(s)
Cell Culture Techniques , Extracellular Matrix , Animals , Basement Membrane , Cell Culture Techniques/methods , Cell Differentiation , Cell Line, Tumor , Extracellular Matrix/metabolism
4.
Nat Commun ; 11(1): 5799, 2020 11 16.
Article in English | MEDLINE | ID: mdl-33199705

ABSTRACT

The extent and importance of functional heterogeneity and crosstalk between tumor cells is poorly understood. Here, we describe the generation of clonal populations from a patient-derived ovarian clear cell carcinoma model which forms malignant ascites and solid peritoneal tumors upon intraperitoneal transplantation in mice. The clonal populations are engineered with secreted Gaussia luciferase to monitor tumor growth dynamics and tagged with a unique DNA barcode to track their fate in multiclonal mixtures during tumor progression. Only one clone, CL31, grows robustly, generating exclusively malignant ascites. However, multiclonal mixtures form large solid peritoneal metastases, populated almost entirely by CL31, suggesting that transient cooperative interclonal interactions are sufficient to promote metastasis of CL31. CL31 uniquely harbors ERBB2 amplification, and its acquired metastatic activity in clonal mixtures is dependent on transient exposure to amphiregulin, which is exclusively secreted by non-tumorigenic clones. Amphiregulin enhances CL31 mesothelial clearance, a prerequisite for metastasis. These findings demonstrate that transient, ostensibly innocuous tumor subpopulations can promote metastases via "hit-and-run" commensal interactions.


Subject(s)
Cell Communication , Clone Cells/pathology , Neoplasm Metastasis/pathology , Amphiregulin/metabolism , Animals , Ascites/pathology , Carcinogenesis/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation , Cell Separation , Cohort Studies , DNA Copy Number Variations/genetics , Epithelium/pathology , Female , Gene Amplification , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Ligands , Mice, SCID , Models, Biological , Peritoneal Neoplasms/secondary , Phenotype , Receptor, ErbB-2/genetics , Time Factors
5.
Elife ; 72018 08 09.
Article in English | MEDLINE | ID: mdl-30091702

ABSTRACT

VivosX is an in vivo disulfide crosslinking approach that utilizes a pair of strategically positioned cysteines on two proteins to probe physical interactions within cells. Histone H2A.Z, which often replaces one or both copies of H2A in nucleosomes downstream of promoters, was used to validate VivosX. Disulfide crosslinks between cysteine-modified H2A.Z and/or H2A histones within nucleosomes were induced using a membrane-permeable oxidant. VivosX detected different combinations of H2A.Z and H2A within nucleosomes in yeast cells. This assay correctly reported the change in global H2A.Z occupancy previously observed when the deposition and eviction pathways of H2A.Z were perturbed. Homotypic H2A.Z/H2A.Z (ZZ) nucleosomes accumulated when assembly of the transcription preinitiation complex was blocked, revealing that the transcription machinery preferentially disassembles ZZ nucleosomes. VivosX works in human cells and distinguishes ZZ nucleosomes with one or two ubiquitin moieties, demonstrating that it can be used to detect protein-protein interactions inside cells from different species.


Subject(s)
Disulfides/metabolism , Histones/metabolism , Protein Interaction Mapping/methods , Cell Line , Humans , Nucleosomes/chemistry , Oxidation-Reduction , Protein Binding , Saccharomyces cerevisiae/chemistry
6.
Nature ; 510(7503): 167-71, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24739973

ABSTRACT

Centrosome amplification has long been recognized as a feature of human tumours; however, its role in tumorigenesis remains unclear. Centrosome amplification is poorly tolerated by non-transformed cells and, in the absence of selection, extra centrosomes are spontaneously lost. Thus, the high frequency of centrosome amplification, particularly in more aggressive tumours, raises the possibility that extra centrosomes could, in some contexts, confer advantageous characteristics that promote tumour progression. Using a three-dimensional model system and other approaches to culture human mammary epithelial cells, we find that centrosome amplification triggers cell invasion. This invasive behaviour is similar to that induced by overexpression of the breast cancer oncogene ERBB2 (ref. 4) and indeed enhances invasiveness triggered by ERBB2. Our data indicate that, through increased centrosomal microtubule nucleation, centrosome amplification increases Rac1 activity, which disrupts normal cell-cell adhesion and promotes invasion. These findings demonstrate that centrosome amplification, a structural alteration of the cytoskeleton, can promote features of malignant transformation.


Subject(s)
Breast Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Centrosome/pathology , Genes, erbB-2 , Aneuploidy , Breast/cytology , Breast/pathology , Breast Neoplasms/genetics , Cell Adhesion , Cell Line , Cell Transformation, Neoplastic/genetics , Disease Progression , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/pathology , Humans , Microtubules/chemistry , Microtubules/metabolism , Microtubules/pathology , Neoplasm Invasiveness/pathology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , rac1 GTP-Binding Protein/metabolism
7.
PLoS One ; 9(3): e90722, 2014.
Article in English | MEDLINE | ID: mdl-24599099

ABSTRACT

The potent tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA) alters many cellular processes through activation of its receptor protein kinase C (PKC), including gene expression, cell cycle, and the regulation of cell morphology, raising an important question for developing targeted methods to prevent cancer: which effects of TPA are crucial for carcinogenesis? To address this question, we studied TPA action in the 3-dimensional (3D) MCF10A human breast epithelial cell system, which models important features of in vivo epithelial tissue including growth constraints, structural organization of cells, and establishment of a basement membrane. MCF10A cells, which are immortalized but nontumorigenic, form hollow, spheroid structures in 3D culture referred to as acini. The development of normal acini requires the tight spatiotemporal regulation of cellular proliferation, polarization, apoptosis, and growth arrest. Treatment of MCF10A acini with TPA caused the appearance of multi-acinar structures. Surprisingly, this phenotype did not involve an increase in cell number or major changes in cell death, and polarization. Instead, live cell and confocal microscopy revealed that TPA stimulates MCF10A acini to aggregate. TPA induces the PKC-dependent production of actin-based protrusions, which leads to the formation of cellular bridges between acini, the clustering of acini, and allows cells to move into adjacent acini. During this process, the integrity of the laminin V basement membrane is disrupted, while E-cadherin-based cell-cell contacts remain intact. Altogether, our results show that under the biochemical and structural constraints of epithelial tissue, as modeled by the 3D MCF10A system, TPA induces a novel PKC-dependent phenotype that resembles local invasion. Of the many effects caused by TPA, these studies highlight the aggressive production of actin-based cellular protrusions as a potentially important event along the pathway to carcinogenesis.


Subject(s)
Acinar Cells/enzymology , Acinar Cells/pathology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carcinogenesis/pathology , Mammary Glands, Human/pathology , Protein Kinase C/metabolism , Actins/metabolism , Cadherins/metabolism , Cell Aggregation/drug effects , Cell Death/drug effects , Cell Line , Cell Polarity/drug effects , Female , Humans , Laminin/metabolism , Mitosis/drug effects , Neoplasm Invasiveness , Phenotype , Tetradecanoylphorbol Acetate , Time Factors
9.
Nature ; 482(7385): 410-3, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22318515

ABSTRACT

Tumorigenesis is a clonal evolution process that is initiated from single cells within otherwise histologically normal tissue. It is unclear how single, sporadic mutant cells that have sustained oncogenic alterations evolve within a tightly regulated tissue environment. Here we investigated the effects of inducing oncogene expression in single cells in organotypic mammary acini as a model to elucidate the processes by which oncogenic alterations initiate clonal progression from organized epithelial environments. Sporadic cells induced to overexpress oncogenes that specifically perturb cell-cycle checkpoints (for example, E7 from human papilloma virus 16, and cyclin D1), deregulate Myc transcription or activate AKT signalling remained quiescent within growth-arrested acini. By contrast, single cells that overexpress ERBB2 initiated a cellular cascade involving cell translocation from the epithelial layer, as well as luminal outgrowth that is characteristic of neoplastic progression in early-stage epithelial tumours. In addition, ERBB2-mediated cell translocation to the lumen was found to depend on extracellular-regulated kinase and matrix metalloproteinase activities, and genetic alterations that perturb local cell-matrix adhesion drove cell translocation. We also provide evidence that luminal cell translocation may drive clonal selection by promoting either the death or the expansion of quiescent oncogene-expressing cells, depending on whether the pre-existing alterations allow anchorage-independent survival and growth. Our data show that the initial outgrowth of single oncogene-expressing cells from organized epithelial structures is a highly regulated process, and we propose that a cell translocation mechanism allows sporadic mutant cells to evade suppressive micro-environments and elicits clonal selection for survival and proliferative expansion outside the native niches of these cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cellular Microenvironment/physiology , Clonal Evolution , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Oncogenes/genetics , Acinar Cells/cytology , Acinar Cells/metabolism , Acinar Cells/pathology , Cell Adhesion , Cell Culture Techniques , Cell Line , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Contact Inhibition , Epithelial Cells/cytology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Microenvironment
10.
Cell ; 139(7): 1226-8, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20064369

ABSTRACT

Circulating tumor cells are responsible for seeding metastatic growth at distant sites. Kim et al. (2009) now discover that circulating tumor cells can reinfiltrate tumors at their primary organs and promote tumor progression.


Subject(s)
Neoplasm Seeding , Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Animals , Disease Progression , Humans , Neoplasms/physiopathology
11.
J Neurosci ; 27(47): 12764-74, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18032648

ABSTRACT

In the adult mammalian brain, new neurons and glia are continuously generated but molecular factors regulating their differentiation and lineage relationships are largely unknown. We show that Ascl1, a bHLH (basic helix-loop-helix) transcription factor, transiently labels neuronal and oligodendrocyte precursors in the adult brain. Using in vivo lineage tracing with inducible Cre recombinase, we followed the maturation of these precursors in four distinct regions. In the hippocampus, Ascl1 mostly marks type-2a progenitor cells with some late stage type-1 stem cells. Thirty days after Ascl1 expression, although a majority of the cells matured to granule neurons, a few cells remained as immature progenitors. By 6 months, however, essentially all Ascl1 lineage cells were granule neurons. In contrast, in the olfactory bulb neuronal lineage, Ascl1 is restricted to transit amplifying cells, and by 30 d all cells matured into GABAergic interneurons. Ascl1 also broadly marks oligodendrocyte precursors in subcortical gray and white matter regions. In the corpus callosum, Ascl1 defines a ventral layer of early oligodendrocyte precursors that do not yet express other early markers of this lineage like PDGFRalpha and Olig2. By 30 d, most had transitioned to mature oligodendrocytes. In contrast, Ascl1 expressing oligodendrocyte precursors in gray matter already coexpressed the early oligodendrocyte markers, but by 30 d they mostly remained as precursors. Our results reveal that Ascl1 is a common molecular marker of early progenitors of both neurons and oligodendrocytes in the adult brain, and these Ascl1 defined progenitors mature with distinct dynamics in different brain regions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/analysis , Cell Differentiation/physiology , Neuroglia/cytology , Neurons/cytology , Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Brain/cytology , Brain/growth & development , Brain/metabolism , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Transgenic , Neuroglia/chemistry , Neuroglia/physiology , Neurons/physiology , Stem Cells/chemistry , Stem Cells/physiology
12.
Nat Neurosci ; 10(6): 720-6, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17468753

ABSTRACT

The olfactory neuroepithelium undergoes continual neurogenesis and, after extensive lesions, fully regenerates to maintain sensory function. The stem cell population underlying this regenerative capacity remains elusive. Here we show that mouse horizontal basal cells (HBCs) function as adult olfactory neuroepithelium neural stem cells and examine their distinct dynamics in olfactory neuroepithelium maintenance and regeneration. Fate-mapping analysis after olfactory neuroepithelium lesioning shows that HBCs are competent to regenerate both neuronal and non-neuronal olfactory neuroepithelium lineages. HBCs serve as a reservoir of long-lived progenitors that remain largely quiescent during normal neuronal turnover or even after acute, selective loss of mature neurons. Under these conditions, previously identified progenitors are largely responsible for tissue maintenance. Yet after extensive injuries that deplete resident neuronal precursors, HBCs transiently proliferate and their progeny fully reconstitute the neuroepithelium. Our data support a new model of adult neurogenesis in which distinct cell populations mediate normal neuronal turnover and neuronal replacement upon traumatic injury.


Subject(s)
Adult Stem Cells/physiology , Neuroepithelial Cells/physiology , Olfactory Bulb/cytology , Olfactory Pathways/physiology , Regeneration/physiology , Animals , Antigens, Bacterial/genetics , Antigens, Surface/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Bromodeoxyuridine/metabolism , Cell Count , Cell Differentiation/physiology , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Olfactory Marker Protein/metabolism , Olfactory Pathways/drug effects , Olfactory Pathways/injuries , Tamoxifen/analogs & derivatives , Tamoxifen/toxicity , Time Factors
13.
J Virol ; 76(1): 78-87, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11739673

ABSTRACT

Mink cell focus-inducing (MCF) viruses induce T-cell lymphomas in AKR/J strain mice. MCF 247, the prototype of this group of nonacute murine leukemia viruses, transforms thymocytes, in part, by insertional mutagenesis and enhancer-mediated dysregulation of cellular proto-oncogenes. The unique 3' (U3) regions in the long terminal repeats of other murine leukemia viruses contain transcription factor binding sites known to be important for enhancer function and for the induction of T-cell lymphomas. Although transcription factor binding sites important for the biological properties of MCF 247 have not been identified, pathogenesis studies from our laboratory suggested to us that binding sites for Ikaros, a lymphoid-cell-restricted transcriptional regulator, affect the biological properties of MCF 247. In this report, we demonstrate that Ikaros binds to predicted sites in U3 sequences of MCF 247 and that site-directed mutations in these sites greatly diminish this binding in vitro. Consistent with these findings, ectopic expression of Ikaros in murine cells that do not normally express this protein significantly increases transcription from the viral promoter in transient gene expression assays. Moreover, site-directed mutations in specific Ikaros-binding sites reduce this activity in T-cell lines that express Ikaros endogenously. To determine whether the Ikaros-binding sites are functional in vivo, we inoculated newborn mice with a variant MCF virus containing a mutant Ikaros-binding site. The variant virus replicated in thymocytes less efficiently and induced lymphomas with a delayed onset compared to the wild-type virus. These data are consistent with the hypothesis that the Ikaros-binding sites in the U3 region of MCF 247 are functional and cooperate with other DNA elements for optimal enhancer function in vivo.


Subject(s)
DNA-Binding Proteins , Mink Cell Focus-Inducing Viruses/pathogenicity , Transcription Factors/physiology , 3' Untranslated Regions/genetics , 3' Untranslated Regions/metabolism , 3T3 Cells , Animals , Animals, Newborn , Binding Sites , Cell Line , Humans , Ikaros Transcription Factor , Jurkat Cells , Leukemia, Experimental/virology , Mice , Mice, Inbred AKR , Mink Cell Focus-Inducing Viruses/genetics , Mutagenesis, Site-Directed , Protein Binding , Retroviridae Infections/virology , Transcription Factors/metabolism , Transcription, Genetic , Tumor Virus Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...