Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where E. Lynette Wilson is active.

Publication


Featured researches published by E. Lynette Wilson.


Journal of Cell Biology | 2002

Proximal location of mouse prostate epithelial stem cells: a model of prostatic homeostasis

Akira Tsujimura; Yasuhiro Koikawa; Sarah N. Salm; Tetsuya Takao; Sandra Coetzee; David Moscatelli; Ellen Shapiro; Herbert Lepor; Tung-Tien Sun; E. Lynette Wilson

Stem cells are believed to regulate normal prostatic homeostasis and to play a role in the etiology of prostate cancer and benign prostatic hyperplasia. We show here that the proximal region of mouse prostatic ducts is enriched in a subpopulation of epithelial cells that exhibit three important attributes of epithelial stem cells: they are slow cycling, possess a high in vitro proliferative potential, and can reconstitute highly branched glandular ductal structures in collagen gels. We propose a model of prostatic homeostasis in which mouse prostatic epithelial stem cells are concentrated in the proximal region of prostatic ducts while the transit-amplifying cells occupy the distal region of the ducts. This model can account for many biological differences between cells of the proximal and distal regions, and has implications for prostatic disease formation.


Journal of Cellular Physiology | 1999

CELLULAR GLYCOSYLPHOSPHATIDYLINOSITOL-SPECIFIC PHOSPHOLIPASE D REGULATES UROKINASE RECEPTOR SHEDDING AND CELL SURFACE EXPRESSION

Olaf Wilhelm; Sabine Wilhelm; Gemma M. Escott; Verena Lutz; Viktor Magdolen; Manfred Schmitt; Daniel B. Rifkin; E. Lynette Wilson; H. Graeff; Georg Brunner

The glycosylphosphatidylinositol (GPI) ‐ anchored, multifunctional receptor for the serine proteinase, urokinase plasminogen activator (uPAR, CD87), regulates plasminogen activation and cell migration, adhesion, and proliferation. uPAR occurs in functionally distinct, membrane‐anchored and soluble isoforms (s‐uPAR) in vitro and in vivo. Recent evidence indicates that s‐uPAR present in the circulation of cancer patients correlates with tumor malignancy and represents a valuable prognostic marker in certain types of cancer. We have therefore analyzed the mechanism of uPAR shedding in vitro. We present evidence that uPAR is actively released from ovarian cancer cells since the rate of receptor shedding did not correlate with uPAR expression. While s‐uPAR was derived from the cell surface, it lacked the hydrophobic portion of the GPI moiety indicating anchor cleavage. We show that uPAR release is catalyzed by cellular GPI‐specific phospholipase D (GPI‐PLD), an enzyme cleaving the GPI anchor of the receptor. Thus, recombinant GPI‐PLD expression increased receptor release up to fourfold. Conversely, a 40% reduction in GPI‐PLD activity by GPI‐PLD antisense mRNA expression inhibited uPAR release by more than 60%. We found that GPI‐PLD also regulated uPAR expression, possibly by releasing a GPI‐anchored growth factor. Our data suggest that cellular GPI‐PLD might be involved in the generation of circulating prognostic markers in cancer and possibly regulate the function of GPI‐anchored proteins by generating functionally distinct, soluble counterparts. J. Cell. Physiol. 180:225–235, 1999.


Stem Cells | 2009

High Aldehyde Dehydrogenase Activity: A Novel Functional Marker of Murine Prostate Stem/Progenitor Cells

Patricia E. Burger; Rashmi Gupta; Xiaozhong Xiong; Christopher S. Ontiveros; Sarah N. Salm; David Moscatelli; E. Lynette Wilson

We have shown previously that prostatic stem/progenitor cells can be purified from isolated prostate ducts, based on their high expression of the Sca‐1 surface antigen. We now report that high levels of aldehyde dehydrogenase (ALDH) activity are present in a subset of prostate epithelial cells that coexpress a number of antigens found on stem/progenitor cells of other origins (CD9, Bcl‐2, CD200, CD24, prominin, Oct 3/4, ABCG2, and nestin). Almost all of these cells expressing high levels of ALDH activity also express Sca‐1 and a third of them express high levels of this antigen. The cells with high levels of ALDH activity have greater in vitro proliferative potential than cells with low ALDH activity. Importantly, in an in vivo prostate reconstitution assay, the cells expressing high levels of ALDH activity were much more effective in generating prostatic tissue than a population of cells with low enzymatic activity. Thus, a high level of ALDH activity can be considered a functional marker of prostate stem/progenitor cells and allows for simple, efficient isolation of cells with primitive features. The elucidation of the role of ALDH in prostate stem/progenitor cells may lead to the development of rational therapies for treating prostate cancer and benign prostatic hyperplasia. STEM CELLS 2009;27:2220–2228


Journal of Cell Biology | 2005

TGF-β maintains dormancy of prostatic stem cells in the proximal region of ducts

Sarah N. Salm; Patricia E. Burger; Sandra Coetzee; Ken Goto; David Moscatelli; E. Lynette Wilson

We have previously shown that prostatic stem cells are located in the proximal region of mouse prostatic ducts. Here, we show that this region responds differently to transforming growth factor (TGF)-β than the distal ductal region and that under physiological conditions androgens and TGF-β are crucial overall regulators of prostatic tissue homeostasis. This conclusion is supported by the observations showing that high levels of TGF-β signaling are present in the quiescent proximal region of ducts in an androgen-replete animal and that cells in this region overexpress Bcl-2, which protects them from apoptosis. Moreover, androgen ablation reverses the proximal-distal TGF-β signaling gradient, leading to an increase in TGF-β signaling in the unprotected distal region (low Bcl-2 expression). This reversal of TGF-β–mediated signaling accompanies apoptosis of cells in the distal region and gland involution after androgen withdrawal. A physiological TGF-β signaling gradient (high proximally and low distally) and its functional correlates are restored after androgen replenishment. In addition to highlighting the regulatory role of androgens and TGF-β, these findings may have important implications for the deregulation of the stem cell compartment in the etiology of proliferative prostatic diseases.


Growth Factors Journal | 1990

Long-term culture of human bone marrow stromal cells in the presence of basic fibroblast growth factor.

Lisa Oliver; Daniel B. Rifkin; Janice Gabrilove; Melanie-Jane Hannocks; E. Lynette Wilson

Basic fibroblast growth factor (bFGF) is a potent mitogen for human bone marrow stromal cells. Normally, large numbers of human bone marrow stromal cells are difficult to obtain. However, nanogram/ml concentrations of bFGF stimulate the growth of passaged bone marrow stromal cells both in media formulated for optimal growth of stromal cells and in a simple mixture of RPMI-1640 and 10% fetal calf serum facilitating the successive expansion of stromal cells through multiple passages. bFGF also greatly accelerates the formation of a primary stromal cell layer following inoculation of newly harvested bone marrow cells into dishes. In the presence of bFGF, the stromal cells attain high densities, lose their contact inhibition and grow in multilayered sheets. Heparin greatly potentiates the stimulatory effect of low concentrations of bFGF. The effects of bFGF are fully reversible: cells cultured in the presence of this factor for multiple passages revert to normal growth rates following trypsinization and subculture. A short (4 h) exposure of the cells to bFGF elicits profound growth stimulation. This supports the hypothesis that this factor binds to glycosaminoglycans in the cell matrix which act as a storage reservoir for this cytokine.


Stem Cells | 2006

Proximal Prostatic Stem Cells Are Programmed to Regenerate a Proximal‐Distal Ductal Axis

Ken Goto; Sarah N. Salm; Sandra Coetzee; Xiaozhong Xiong; Patricia E. Burger; Ellen Shapiro; Herbert Lepor; David Moscatelli; E. Lynette Wilson

Prostate carcinoma and benign prostatic hypertrophy may both originate in stem cells, highlighting the importance of the characterization of these cells. The prostate gland contains a network of ducts each of which consists of a proximal (adjacent to the urethra), an intermediate, and a distal region. Here, we report that two populations of cells capable of regenerating prostatic tissue in an in vivo prostate reconstitution assay are present in different regions of prostatic ducts. The first population (with considerable growth potential) resides in the proximal region of ducts and in the urethra, and the survival of these cells does not require the presence of androgens. The second population (with more limited growth potential) is found in the remaining ductal regions and requires androgen for survival. In addition, we find that primitive proximal prostate cells that are able to regenerate functional prostatic tissue in vivo are also programmed to re‐establish a proximal‐distal ductal axis. Similar to their localization in the intact prostate, cells with the highest regenerative capacity are found in the proximal region of prostatic ducts formed in an in vivo prostate reconstitution assay. The primitive proximal cells can be passaged through four generations of subrenal capsule grafts. Together, these novel findings illustrate features of primitive prostate cells that may have implications for the development of therapies for treating proliferative prostatic diseases.


PLOS ONE | 2009

Molecular Signatures of Prostate Stem Cells Reveal Novel Signaling Pathways and Provide Insights into Prostate Cancer

Roy Blum; Rashmi Gupta; Patricia E. Burger; Christopher S. Ontiveros; Sarah N. Salm; Xiaozhong Xiong; Alexander Kamb; Holger Wesche; Lisa Marshall; Gene Cutler; Xiangyun Wang; Jiri Zavadil; David Moscatelli; E. Lynette Wilson

Background The global gene expression profiles of adult and fetal murine prostate stem cells were determined to define common and unique regulators whose misexpression might play a role in the development of prostate cancer. Methodology/Principal Findings A distinctive core of transcriptional regulators common to both fetal and adult primitive prostate cells was identified as well as molecules that are exclusive to each population. Elements common to fetal and adult prostate stem cells include expression profiles of Wnt, Shh and other pathways identified in stem cells of other organs, signatures of the aryl-hydrocarbon receptor, and up-regulation of components of the aldehyde dehydrogenase/retinoic acid receptor axis. There is also a significant lipid metabolism signature, marked by overexpression of lipid metabolizing enzymes and the presence of the binding motif for Srebp1. The fetal stem cell population, characterized by more rapid proliferation and self-renewal, expresses regulators of the cell cycle, such as E2f, Nfy, Tead2 and Ap2, at elevated levels, while adult stem cells show a signature in which TGF-β has a prominent role. Finally, comparison of the signatures of primitive prostate cells with previously described profiles of human prostate tumors identified stem cell molecules and pathways with deregulated expression in prostate tumors including chromatin modifiers and the oncogene, Erg. Conclusions/Significance Our data indicate that adult prostate stem or progenitor cells may acquire characteristics of self-renewing primitive fetal prostate cells during oncogenesis and suggest that aberrant activation of components of prostate stem cell pathways may contribute to the development of prostate tumors.


Journal of Cellular Physiology | 2000

Generation of active TGF-β by prostatic cell cocultures using novel basal and luminal prostatic epithelial cell lines

Sarah N. Salm; Yasuhiro Koikawa; Varrie Ogilvie; Akira Tsujimura; Sandra Coetzee; David Moscatelli; Emma E. Moore; Herbert Lepor; Ellen Shapiro; Tung-Tien Sun; E. Lynette Wilson

Two prostatic epithelial lines, one of basal origin and one of luminal origin, were established from the dorsolateral prostates of p53 null mice. The cell lines are nontumorigenic when inoculated subcutaneously under the renal capsule or intraprostatically in syngeneic mice. The luminal cell line (PE‐L‐1) expresses cytokeratins 8 and 18 and the basal cell line (PE‐B‐1) expresses cytokeratins 5 and 14. The basal cells require serum for growth, whereas the luminal cells grow only in serum‐free medium. Both cell lines require the presence of growth factors for optimal growth in culture, with EGF and FGF‐2 having the greatest effect on the growth rate. Both lines express androgen receptor (AR) mRNA and protein. Androgen stimulates growth of the basal cell line, indicating that the ARs are functional, whereas growth of the luminal cells is unaffected by androgens. The luminal line is significantly inhibited by exogenous TGF‐β and produces low levels of endogenous TGF‐β. In contrast, the basal cell line produces significant amounts of TGF‐β and its growth is not influenced by this cytokine. Coculture of luminal cells with prostatic smooth muscle cells results in the generation of increased levels of biologically active TGF‐β, indicating a paracrine mechanism of TGF‐β activation that may be involved in the maintenance of normal prostatic function. To our knowledge this is the first report describing both basal and luminal prostatic cell lines from a single inbred animal species and the first indication that prostatic epithelial and stromal cells interact to generate the biologically active form of TGF‐β. These lines will provide an important model for determining basal/luminal interactions in both in vitro and in vivo assays. J. Cell. Physiol. 184:70–79, 2000.


Journal of Cellular Physiology | 2000

Transforming growth factor-β is an autocrine mitogen for a novel androgen-responsive murine prostatic smooth muscle cell line, PSMC1

Sarah N. Salm; Yasuhiro Koikawa; Varrie Ogilvie; Akira Tsujimura; Sandra Coetzee; David Moscatelli; Emma E. Moore; Herbert Lepor; Ellen Shapiro; Tung-Tien Sun; E. Lynette Wilson

A prostatic smooth muscle cell line (PSMC1) was established from the dorsolateral prostate of p53 null mice. The cell line is nontumorigenic when inoculated subcutaneously, under the renal capsule or intraprostatically in syngeneic mice. These cells express α–smooth muscle actin (α‐SMA), indicating their smooth muscle origin, and TGF‐β significantly enhances expression of α‐SMA. The cells express both androgen receptor (AR) mRNA and protein, and respond mitogenically to physiological concentrations of androgens. PSMC1 cells produce significant amounts of TGF‐β, which stimulates growth by an autocrine mechanism. Dihydrotestosterone (DHT) increases proliferation of PSMC1 cells by promoting TGF‐β secretion. Considering the significant inhibitory effect of TGF‐β on prostatic epithelial cells and its stimulatory effect on the PSMC1 cells, we postulate that TGF‐β produced by prostatic smooth muscle cells may have a paracrine effect on the prostatic epithelium. We also postulate that TGF‐β may be involved in the etiology of benign prostatic hyperplasia (BPH) by stimulating excessive stromal proliferation. Line PSMC1 is the first reported androgen‐responsive murine smooth muscle cell line. It will be useful for in vivo and in vitro experiments to study the mechanisms of androgen action on prostatic stroma and for delineating the interactions that occur between prostatic smooth muscle and epithelium that may lead to prostatic diseases such as BPH. J. Cell. Physiol. 185:416–424, 2000.


PLOS ONE | 2010

Molecular signatures of the primitive prostate stem cell niche reveal novel mesenchymal-epithelial signaling pathways.

Roy Blum; Rashmi Gupta; Patricia E. Burger; Christopher S. Ontiveros; Sarah N. Salm; Xiaozhong Xiong; Alexander Kamb; Holger Wesche; Lisa Marshall; Gene Cutler; Xiangyun Wang; Jiri Zavadil; David Moscatelli; E. Lynette Wilson

Background Signals between stem cells and stroma are important in establishing the stem cell niche. However, very little is known about the regulation of any mammalian stem cell niche as pure isolates of stem cells and their adjacent mesenchyme are not readily available. The prostate offers a unique model to study signals between stem cells and their adjacent stroma as in the embryonic prostate stem cell niche, the urogenital sinus mesenchyme is easily separated from the epithelial stem cells. Here we investigate the distinctive molecular signals of these two stem cell compartments in a mammalian system. Methodology/Principal Findings We isolated fetal murine urogenital sinus epithelium and urogenital sinus mesenchyme and determined their differentially expressed genes. To distinguish transcripts that are shared by other developing epithelial/mesenchymal compartments from those that pertain to the prostate stem cell niche, we also determined the global gene expression of epidermis and dermis of the same embryos. Our analysis indicates that several of the key transcriptional components that are predicted to be active in the embryonic prostate stem cell niche regulate processes such as self-renewal (e.g., E2f and Ap2), lipid metabolism (e.g., Srebp1) and cell migration (e.g., Areb6 and Rreb1). Several of the enriched promoter binding motifs are shared between the prostate epithelial/mesenchymal compartments and their epidermis/dermis counterparts, indicating their likely relevance in epithelial/mesenchymal signaling in primitive cellular compartments. Based on differential gene expression we also defined ligand-receptor interactions that may be part of the molecular interplay of the embryonic prostate stem cell niche. Conclusions/Significance We provide a comprehensive description of the transcriptional program of the major regulators that are likely to control the cellular interactions in the embryonic prostatic stem cell niche, many of which may be common to mammalian niches in general. This study provides a comprehensive source for further studies of mesenchymal/epithelial interactions in the prostate stem cell niche. The elucidation of pathways in the normal primitive niche may provide greater insight into mechanisms subverted during abnormal proliferative and oncogenic processes. Understanding these events may result in the development of specific targeted therapies for prostatic diseases such as benign prostatic hypertrophy and carcinomas.

Collaboration


Dive into the E. Lynette Wilson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge