Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 7180, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893591

ABSTRACT

Metastasis is the main cause of carcinoma-related death, yet we know little about how it initiates due to our inability to visualize stochastic invasion events. Classical models suggest that cells accumulate mutations that first drive formation of a primary mass, and then downregulate epithelia-specific genes to cause invasion and metastasis. Here, using transparent zebrafish epidermis to model simple epithelia, we can directly image invasion. We find that KRas-transformation, implicated in early carcinogenesis steps, directly drives cell invasion by hijacking a process epithelia normally use to promote death-cell extrusion. Cells invading by basal cell extrusion simultaneously pinch off their apical epithelial determinants, endowing new plasticity. Following invasion, cells divide, enter the bloodstream, and differentiate into stromal, neuronal-like, and other cell types. Yet, only invading KRasV12 cells deficient in p53 survive and form internal masses. Together, we demonstrate that KRas-transformation alone causes cell invasion and partial dedifferentiation, independently of mass formation.


Subject(s)
Epithelial Cells/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Movement , Epidermis/metabolism , Epithelium/metabolism , Humans , Neoplasms/diagnostic imaging , Zebrafish/metabolism , Zebrafish Proteins
2.
J Cell Sci ; 130(1): 269-277, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27149923

ABSTRACT

Epithelia provide a crucial protective barrier for our organs and are also the sites where the majority of carcinomas form. Most studies on epithelia and carcinomas use cell culture or organisms where high-resolution live imaging is inaccessible without invasive techniques. Here, we introduce the developing zebrafish epidermis as an excellent in vivo model system for studying a living epithelium. We developed tools to fluorescently tag specific epithelial cell types and express genes in a mosaic fashion using five Gal4 lines identified from an enhancer trap screen. When crossed to a variety of UAS effector lines, we can now track, ablate or monitor single cells at sub-cellular resolution. Using photo-cleavable morpholino oligonucleotides that target gal4, we can also express genes in a mosaic fashion at specific times during development. Together, this system provides an excellent in vivo alternative to tissue culture cells, without the intrinsic concerns of culture conditions or transformation, and enables the investigation of distinct cell types within living epithelial tissues.


Subject(s)
Cytological Techniques/methods , Epidermal Cells , Zebrafish/metabolism , Animals , Cell Death/drug effects , Cell Division/drug effects , Crosses, Genetic , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Epidermis/drug effects , Epidermis/ultrastructure , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Imaging, Three-Dimensional , Male , Morpholinos/pharmacology , Time Factors , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/metabolism
3.
Elife ; 4: e04069, 2015 Jan 26.
Article in English | MEDLINE | ID: mdl-25621765

ABSTRACT

When epithelia become too crowded, some cells are extruded that later die. To extrude, a cell produces the lipid, Sphingosine 1-Phosphate (S1P), which activates S1P2 receptors in neighboring cells that seamlessly squeeze the cell out of the epithelium. Here, we find that extrusion defects can contribute to carcinogenesis and tumor progression. Tumors or epithelia lacking S1P2 cannot extrude cells apically and instead form apoptotic-resistant masses, possess poor barrier function, and shift extrusion basally beneath the epithelium, providing a potential mechanism for cell invasion. Exogenous S1P2 expression is sufficient to rescue apical extrusion, cell death, and reduce orthotopic pancreatic tumors and their metastases. Focal Adhesion Kinase (FAK) inhibitor can bypass extrusion defects and could, therefore, target pancreatic, lung, and colon tumors that lack S1P2 without affecting wild-type tissue.


Subject(s)
Cell Polarity , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Aggregation/drug effects , Cell Line, Tumor , Cell Polarity/drug effects , Dogs , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/pathology , Epidermis/drug effects , Epidermis/embryology , Epidermis/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Madin Darby Canine Kidney Cells , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Kinase Inhibitors/pharmacology , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Zebrafish/embryology
4.
Nat Rev Cancer ; 14(7): 495-501, 2014 07.
Article in English | MEDLINE | ID: mdl-24943812

ABSTRACT

Metastasis is the leading cause of cancer-related deaths, but it is unclear how cancer cells escape their primary sites in epithelia and disseminate to other sites in the body. One emerging possibility is that transformed epithelial cells could invade the underlying tissue by a process called cell extrusion, which epithelia use to remove cells without disrupting their barrier function. Typically, during normal cell turnover, live cells extrude apically from the epithelium into the lumen and later die by anoikis; however, several oncogenic mutations shift cell extrusion basally, towards the tissue that the epithelium encases. Tumour cells with high levels of survival and motility signals could use basal extrusion to escape from the tissue and migrate to other sites within the body.


Subject(s)
Epithelial Cells/cytology , Neoplasms/physiopathology , Cell Movement , Humans , Neoplasm Invasiveness , Neoplasms/pathology
5.
Curr Biol ; 24(1): 19-28, 2014 Jan 06.
Article in English | MEDLINE | ID: mdl-24361067

ABSTRACT

BACKGROUND: To maintain a protective barrier, epithelia extrude cells destined to die by contracting a band of actin and myosin. Although extrusion can remove cells triggered to die by apoptotic stimuli, to maintain constant cell numbers, epithelia extrude live cells, which later die by anoikis. Because transformed cells may override anoikis and survive after extrusion, the direction of extrusion has important consequences for the extruded cell's fate. As most cells extrude apically, they are typically eliminated through the lumen; however, cells with upregulated survival signals that extrude basally could potentially invade the underlying tissue and migrate to other sites in the body. RESULTS: We found that oncogenic K-Ras cells predominantly extrude basally, rather than apically, in a cell-autonomous manner and can survive and proliferate after extrusion. Expression of K-Ras(V12) downregulates the bioactive lipid sphingosine 1-phosphate (S1P) and its receptor S1P2, both of which are required for apical extrusion. Surprisingly, the S1P biosynthetic pathway is not affected because the S1P precursor, sphingosine kinase, and the degradative enzymes S1P lyase and S1PP phosphatase are not significantly altered. Instead, we found that high levels of autophagy in extruding Ras(V12) cells leads to S1P degradation. Disruption of autophagy chemically or genetically in K-Ras(V12) cells rescues S1P localization and apical extrusion. CONCLUSIONS: Oncogenic K-Ras cells downregulate both S1P and its receptor S1P2 to promote basal extrusion. Because live basally extruding cells can survive and proliferate after extrusion, we propose that basal cell extrusion provides a novel mechanism for cells to exit the epithelium and initiate invasion into the surrounding tissues.


Subject(s)
Autophagy , Epithelial Cells/metabolism , Lysophospholipids/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Line , Cell Proliferation , Dogs , Epithelial Cells/cytology , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/metabolism
6.
J Cell Biol ; 186(5): 693-702, 2009 Sep 07.
Article in English | MEDLINE | ID: mdl-19720875

ABSTRACT

To preserve epithelial barrier function, dying cells are squeezed out of an epithelium by "apoptotic cell extrusion." Specifically, a cell destined for apoptosis signals its live neighboring epithelial cells to form and contract a ring of actin and myosin II that squeezes the dying cell out of the epithelial sheet. Although most apoptotic cells extrude apically, we find that some exit basally. Localization of actin and myosin IIA contraction dictates the extrusion direction: basal extrusion requires circumferential contraction of neighboring cells at their apices, whereas apical extrusion also requires downward contraction along the basolateral surfaces. To activate actin/myosin basolaterally, microtubules in neighboring cells reorient and target p115 RhoGEF to this site. Preventing microtubule reorientation restricts contraction to the apex, driving extrusion basally. Extrusion polarity has important implications for tumors where apoptosis is blocked but extrusion is not, as basal extrusion could enable these cells to initiate metastasis.


Subject(s)
Apoptosis/physiology , Epithelial Cells/metabolism , Epithelium/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Microtubules/metabolism , Actins/metabolism , Animals , Biomarkers/metabolism , Cell Line , Cell Polarity , Dogs , Epithelial Cells/cytology , Epithelial Cells/drug effects , Guanine Nucleotide Exchange Factors/genetics , Humans , Myosin Type II/metabolism , Nocodazole/pharmacology , Paclitaxel/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rho Guanine Nucleotide Exchange Factors , Tubulin Modulators/pharmacology , Zebrafish/anatomy & histology , Zebrafish/growth & development , Zebrafish/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...