Differentiation of a highly tumorigenic basal cell compartment in urothelial carcinoma.

TitleDifferentiation of a highly tumorigenic basal cell compartment in urothelial carcinoma.
Publication TypeJournal Article
Year of Publication2009
AuthorsHe X, Marchionni L, Hansel DE, Yu W, Sood A, Yang J, Parmigiani G, Matsui W, Berman DM
JournalStem Cells
Volume27
Issue7
Pagination1487-95
Date Published2009 Jul
ISSN1549-4918
KeywordsAged, Animals, Antigens, CD, Carcinoma, Transitional Cell, Cell Adhesion Molecules, Female, GPI-Linked Proteins, Humans, Immunohistochemistry, Keratin-17, Mice, Mice, Nude, Receptors, Laminin, Reverse Transcriptase Polymerase Chain Reaction, Stem Cells, Urinary Bladder Neoplasms, Urothelium, Xenograft Model Antitumor Assays
Abstract

Highly tumorigenic cancer cell (HTC) populations have been identified for a variety of solid tumors and assigned stem cell properties. Strategies for identifying HTCs in solid tumors have been primarily empirical rather than rational, particularly in epithelial tumors, which are responsible for 80% of cancer deaths. We report evidence for a spatially restricted bladder epithelial (urothelial) differentiation program in primary urothelial cancers (UCs) and in UC xenografts. We identified a highly tumorigenic UC cell compartment that resembles benign urothelial stem cells (basal cells), co-expresses the 67-kDa laminin receptor and the basal cell-specific cytokeratin CK17, and lacks the carcinoembryonic antigen family member CEACAM6 (CD66c). This multipotent compartment resides at the tumor-stroma interface, is easily identified on histologic sections, and possesses most, if not all, of the engraftable tumor-forming ability in the parental xenograft. We analyzed differential expression of genes and pathways in basal-like cells versus more differentiated cells. Among these, we found significant enrichment of pathways comprising "hallmarks" of cancer, and pharmacologically targetable signaling pathways, including Janus kinase-signal transducer and activator of transcription, Notch, focal adhesion, mammalian target of rapamycin, epidermal growth factor receptor (erythroblastic leukemia viral oncogene homolog [ErbB]), and wingless-type MMTV integration site family (Wnt). The basal/HTC gene expression signature was essentially invisible within the context of nontumorigenic cell gene expression and overlapped significantly with genes driving progression and death in primary human UC. The spatially restricted epithelial differentiation program described here represents a conceptual advance in understanding cellular heterogeneity of carcinomas and identifies basal-like HTCs as attractive targets for cancer therapy.

DOI10.1002/stem.92
Alternate JournalStem Cells
PubMed ID19544456
Grant ListP50CA088843 / CA / NCI NIH HHS / United States
R01DK072000 / DK / NIDDK NIH HHS / United States
P01CA077664 / CA / NCI NIH HHS / United States
1U54RR023561-01A1 / RR / NCRR NIH HHS / United States
K08DK059375 / DK / NIDDK NIH HHS / United States
Related Faculty: 
Luigi Marchionni, M.D., Ph.D.

Pathology & Laboratory Medicine 1300 York Avenue New York, NY 10065 Phone: (212) 746-6464
Surgical Pathology: (212) 746-2700