Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS One ; 10(5): e0128489, 2015.
Article in English | MEDLINE | ID: mdl-26024527

ABSTRACT

There is evidence that stem cells and their progeny play a role in the development of the prostate. Although stem cells are also considered to give rise to differentiated progeny in the adult prostate epithelium ex vivo, the cohort of adult prostate stem cells in vivo as well as the mechanisms by which the adult prostate epithelium is maintained and regenerated remain highly controversial. We have attempted to resolve this conundrum by performing in vivo tracing of serially replicating cells after the sequential administration of two thymidine analogues to mice. Our results show that, during normal prostate homeostasis, sequentially proliferating cells are detected at a rate that is consistent with a stochastic process. These findings indicate that in vivo, under steady-state conditions, most adult prostate epithelial cells do not represent the progeny of a small number of specialized progenitors that generate sequentially replicating transit-amplifying (TA) cells but are formed by stochastic cell division. Similarly, no rapidly cycling TA cells were detected during regeneration following one cycle of androgen-mediated involution/regeneration of the prostate epithelium. These findings greatly enhance our understanding of the mechanisms regulating prostate epithelial cell renewal and may have significant implications in defining the cell of origin of proliferative prostatic diseases.


Subject(s)
Cell Proliferation/physiology , Prostate/cytology , Stem Cells/cytology , Animals , Epithelium/metabolism , Male , Mice , Prostate/metabolism , Stem Cells/metabolism
2.
Proc Natl Acad Sci U S A ; 110(51): 20611-6, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24218555

ABSTRACT

The adult mouse prostate has a seemingly endless capacity for regeneration, and sonic hedgehog (SHH) signaling has been implicated in this stem cell-driven process. However, it is not clear whether SHH acts on the epithelium or stromal cells that secrete factors required for epithelial expansion. Because little is known about stromal stem cells compared with their epithelial counterparts, we used in vivo mouse genetics tools to characterize four prostate stromal subtypes and their stem cells. Using knockin reporter alleles, we uncovered that SHH signals from prostate basal epithelial cells to adjacent stromal cells. Furthermore, the SHH target gene Gli1 is preferentially expressed in subepithelial fibroblast-like cells, one of four prostate stromal subtypes and the subtype closest to the epithelial source of SHH. Using Genetic Inducible Fate Mapping to mark adult Gli1- or Smooth muscle actin-expressing cells and follow their fate during regeneration, we uncovered that Gli1-expressing cells exhibit long-term self-renewal capacity during multiple rounds of androgen-mediated regeneration after castration-induced involution, and depleted smooth muscle cells are mainly replenished by preexisting smooth muscle cells. Based on our Genetic Inducible Fate Mapping studies, we propose a model where SHH signals to multiple stromal stem cells, which are largely unipotent in vivo.


Subject(s)
Hedgehog Proteins/metabolism , Models, Biological , Prostate/metabolism , Regeneration/physiology , Signal Transduction/physiology , Stem Cells/metabolism , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hedgehog Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Prostate/cytology , Stem Cells/cytology , Stromal Cells/cytology , Stromal Cells/metabolism , Zinc Finger Protein GLI1
3.
PLoS One ; 5(9)2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20941365

ABSTRACT

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.


Subject(s)
Cell Communication , Epithelial Cells/metabolism , Mesoderm/metabolism , Prostate/metabolism , Signal Transduction , Stem Cell Niche/metabolism , Animals , Male , Mesoderm/cytology , Mesoderm/embryology , Mice , Mice, Inbred C57BL , Prostate/cytology , Prostate/embryology , Stem Cell Niche/cytology , Stem Cell Niche/embryology
4.
Sci Transl Med ; 2(43): 43ps38, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20686176

ABSTRACT

The epithelium that lines the surface of prostate glands contains several cell types, including luminal secretory cells and basal cells of unclear function. Despite the fact that prostate tumors contain cells with a luminal phenotype and lack basal cells, a recent report indicates that the cell of origin for human prostate cancer is a basal cell and not a luminal cell. In contrast, another study indicates the reverse. It is possible that both basal and luminal stem/progenitor cells may independently give rise to prostate cancer; a comparison of the molecular signatures of the target cells of transformation with those of prostate tumors may aid in predicting the phenotypes of tumors with aggressive characteristics.


Subject(s)
Cell Lineage , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Humans , Male
5.
Stem Cells ; 27(9): 2220-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19544409

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Prostate/cytology , Prostate/enzymology , Stem Cells/cytology , Stem Cells/enzymology , Animals , Cell Proliferation , Flow Cytometry , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Rats
6.
PLoS One ; 4(5): e5722, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19478945

ABSTRACT

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-beta 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.


Subject(s)
Prostate/cytology , Prostate/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction , Stem Cells/metabolism , Adult , Animals , Cell Proliferation , Fetus/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred C57BL , Models, Biological , Multigene Family , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
7.
Prostate ; 68(12): 1263-72, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18563716

ABSTRACT

BACKGROUND: We previously reported that prostatic stem/progenitor cells are concentrated in the proximal region of prostatic ducts and express stem cell antigen 1 (Sca-1). As Wnt signaling is important for the maintenance of stem cells, we determined whether Sca-1 expressing cells also express Axin2, as Axin2 expression is highly suggestive of active Wnt signaling. METHODS: Axin2 promoter reporter mice were used for whole mount and fluorescence activated cell sorting (FACS) analysis to determine its expression in the prostate. Axin2 expressing cells were also examined for the co-expression of Sca-1. We also used a chemical activator of Wnt signaling, BIO, to determine the effects of Wnt signaling on the growth of primary prostate cells in vitro. RESULTS: We show that Axin2 expression is present in all lobes and is regulated by androgens with the highest Axin2 expression in the lateral and dorsal prostate. Furthermore, a fraction of Axin2 expressing cells co-express Sca-1, suggesting that some progenitor cells have active Wnt signaling. Lastly, we demonstrate that activation of the Wnt pathway may result in increased growth, consistent with a role for Wnt signaling in maintenance and/or expansion of the progenitor cell population. CONCLUSION: Axin2 expressing cells that co-express Sca-1 are present in all prostate lobes suggesting that progenitor cells reside within the Wnt active population. An understanding of the basic biology of signaling pathways mediating growth in the prostate may lead to rational therapies to treat benign prostatic hyperplasia and prostate cancer.


Subject(s)
Cytoskeletal Proteins/metabolism , Prostate/metabolism , Stem Cells/metabolism , Androgens/metabolism , Animals , Antigens, Ly/metabolism , Axin Protein , Cell Proliferation , Cells, Cultured , Epithelial Cells/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Prostate/cytology , Signal Transduction/physiology , Stem Cells/cytology , Wnt Proteins/metabolism
8.
Prostate ; 68(8): 893-901, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18361413

ABSTRACT

INTRODUCTION: The contribution of vascular endothelial cells to prostate growth has not been investigated. We examined whether endothelial cells support growth of prostate tissue when co-inoculated with prostate epithelial cells under the renal capsule. METHODS: Vascular endothelial cells were isolated from mice and co-inoculated under the renal capsule with a prostate luminal or basal epithelial cell line. After 60 days, kidneys were examined for growth of prostate tissue. Prostatic tissues were examined by immunohistochemistry for expression of cytokeratins 5 and 8, and vascular density was determined. To determine if increased expression of VEGF-A would increase prostatic growth, transfected endothelial cells overexpressing VEGF-A were co-inoculated with the prostate luminal or basal epithelial lines. RESULTS: Co-inoculation of endothelial cells and prostate luminal or basal epithelial cells resulted in significant growth of prostatic tissue, whereas inoculation of any of the cell lines alone resulted in little growth. The growths from co-inoculation of endothelial cells and luminal epithelial cells contained duct-like structures that stained with antibodies to cytokeratin 8, whereas those from co-inoculation of endothelial cells and basal epithelial cells contained cords of cells that stained with antibodies to cytokeratin 5. Overexpression of VEGF-A had no effect on growth of the prostatic tissues. CONCLUSION: Endothelial cells contribute to the growth of prostatic epithelial cells.


Subject(s)
Endothelial Cells/cytology , Endothelium, Vascular/cytology , Prostate/cytology , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Male , Mice , Prostate/blood supply , Prostate/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
9.
Prostate ; 67(9): 968-75, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17440972

ABSTRACT

BACKGROUND: The proximal region of the prostatic ducts harbor the prostatic epithelial stem cells. As stem cell niches in other organs are highly vascularized, we determined if the proximal region is more highly vascularized than the remaining regions of the prostate. The effect of androgen on vascular density in the different prostatic regions was also examined. METHODS: Sections from prostates were immunostained with antibodies to CD31, and the vascular density in proximal, intermediate, and distal regions was calculated by image analysis software. Vascular density was compared in prostates from castrated mice that received daily inoculations of testosterone or vehicle alone for 3 days. To examine the role of angiogenic factors in the response to androgen, some animals were also treated with soluble VEGF receptor-2-Fc or Tie-2--Fc fusion proteins, which inhibit the activities of VEGF and angiopoietins, respectively. The endothelial proliferative response to androgen was determined by double staining sections with antibodies to CD31 and Ki-67. RESULTS: In prostates from intact mice, vascular density was highest in the proximal region and lowest in the distal region. Administration of testosterone to castrated mice increased vascular density to the greatest extent in the distal and intermediate regions. The increase in vascular density required VEGF and the angiopoietins. Endothelial cell proliferation was less sensitive to androgen in the proximal region than the remainder of the prostate. CONCLUSIONS: Vascular density is highest in the proximal region of the prostate, but the proximal vessels are less responsive to testosterone.


Subject(s)
Prostate/anatomy & histology , Prostate/blood supply , Regional Blood Flow , Angiogenic Proteins/metabolism , Animals , Blood Flow Velocity , Immunohistochemistry , Male , Mice , Mice, Nude , Orchiectomy , Receptors, Cell Surface/metabolism
10.
Stem Cells ; 24(8): 1859-68, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16644920

ABSTRACT

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.


Subject(s)
Prostate/cytology , Prostate/growth & development , Regeneration/physiology , Stem Cells/cytology , Androgens/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Models, Biological , Rats , Rats, Sprague-Dawley , Regeneration/drug effects , Stem Cells/drug effects , Subrenal Capsule Assay
11.
J Urol ; 174(5): 2051-3, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16217392

ABSTRACT

PURPOSE: We examined the immunolocalization of estrogen receptor (ER)alpha and ERbeta in the human fetal prostate. MATERIALS AND METHODS: Tissue sections from human fetal prostates at 7 to 22 weeks of gestation were stained with antibodies to ERalpha, ERbeta, and cytokeratin 10 and 14. RESULTS: ERalpha expression was not detected until 15 weeks of gestation with sparse staining in the utricle. By 19 weeks increased ERalpha expression was seen in the luminal cells of the ventral urogenital epithelium (UGE), basal cells of the dorsal UGE, utricle, distal periurethral ducts, peripheral stroma and posterior prostatic duct. K14 was detected in basal cells of the UGE and in several posterior acini. At 22 weeks ERalpha expression was more intense in all of these areas. ERbeta was expressed throughout the UGE, ejaculatory ducts, müllerian ducts and entire stroma at 7 weeks. Intense ERbeta staining was observed in these areas and in the prostatic buds by 8 weeks with persistent intense staining through 22 weeks. CONCLUSIONS: To our knowledge we report the first immunolocalization of ERalpha in the human fetal prostate and the earliest demonstration of ERbeta expression in the prostate at 7 weeks of gestation. ERbeta expression is intense during ductal morphogenesis, suggesting a role in normal glandular growth and proliferation. The induction of squamous metaplasia in the UGE, distal periurethral ducts and utricle is associated with ERalpha expression in these areas, while the induction of squamous metaplasia in peripheral prostatic acini is associated with peripheral stromal ERalpha expression. This study suggests estrogen signaling pathways in the human fetal prostate via ERalpha that involve epithelial-epithelial and epithelial-stromal interactions.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Fetus/metabolism , Fetus/pathology , Prostate/pathology , Biopsy, Needle , Estrogen Receptor alpha/analysis , Estrogen Receptor beta/analysis , Humans , Immunohistochemistry , Male , Prostate/immunology , Sensitivity and Specificity , Tissue Culture Techniques
12.
J Cell Biol ; 170(1): 81-90, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15983059

ABSTRACT

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)-beta than the distal ductal region and that under physiological conditions androgens and TGF-beta are crucial overall regulators of prostatic tissue homeostasis. This conclusion is supported by the observations showing that high levels of TGF-beta 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-beta signaling gradient, leading to an increase in TGF-beta signaling in the unprotected distal region (low Bcl-2 expression). This reversal of TGF-beta-mediated signaling accompanies apoptosis of cells in the distal region and gland involution after androgen withdrawal. A physiological TGF-beta 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-beta, these findings may have important implications for the deregulation of the stem cell compartment in the etiology of proliferative prostatic diseases.


Subject(s)
Cell Differentiation/physiology , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Prostate/metabolism , Stem Cells/metabolism , Transforming Growth Factor beta/physiology , Androgens/metabolism , Animals , Apoptosis/physiology , Carcinoma/metabolism , Carcinoma/physiopathology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Male , Mice , Mice, Inbred C57BL , Prostate/cytology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/drug effects , Transforming Growth Factor beta/pharmacology
13.
Proc Natl Acad Sci U S A ; 102(20): 7180-5, 2005 May 17.
Article in English | MEDLINE | ID: mdl-15899981

ABSTRACT

We previously showed that prostatic stem cells are concentrated in the proximal regions of prostatic ducts. We now report that these stem cells can be purified from isolated proximal duct regions by virtue of their high expression of the cell surface protein stem cell antigen 1 (Sca-1). In an in vivo prostate reconstitution assay, the purified Sca-1-expressing cell population isolated from the proximal region of ducts was more effective in generating prostatic tissue than a comparable population of Sca-1-depleted cells (203.0 +/- 83.1 mg vs. 11.9 +/- 9.2 mg) or a population of Sca-1-expressing cells isolated from the remaining regions of ducts (transit-amplifying cells) (31.9 +/- 24.1 mg). Almost all of the proliferative capacity of the proximal duct Sca-1-expressing cell population resides within the fraction of cells that express high levels of Sca-1 (top one-third), with the proximal region of prostatic ducts containing 7.2-fold more Sca-1(high) cells than the remaining regions. More than 60% of the high-expressing cells coexpress alpha6 integrin and the anti-apoptotic factor Bcl-2, markers that are also characteristic of stem cells of other origins. Further stratification of the phenotype of the stem cells may enable the development of rational therapies for treating prostate cancer and benign prostatic hyperplasia.


Subject(s)
Cell Differentiation/physiology , Gene Expression , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Prostate/cytology , Stem Cells/metabolism , Animals , Ataxin-1 , Ataxins , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Rats , Stem Cell Transplantation
14.
Prostate ; 54(1): 17-24, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12481251

ABSTRACT

BACKGROUND: One of the major constraints in elucidating the mechanisms involved in the etiology of benign prostatic hyperplasia (BPH) is the lack of suitable model systems that are readily manipulable in vitro and in vivo. To address this issue, we have used murine prostatic cell lines to establish a novel in vivo model for studying prostatic cell interactions. METHODS: Luminal, basal, and smooth muscle (SM) cell lines were inoculated alone or in combinations under the renal capsule of intact or castrated male mice, and the growth and composition of prostatic tissue in the absence or presence of doxazosin was determined. RESULTS: Both the luminal and basal cell lines reconstituted prostatic tissue if co-inoculated under the renal capsule with normal SM cells, whereas none of the lines formed significant tissue when inoculated alone. Luminal cells produced and secreted prostatic secretory products. The growth of prostatic tissue formed from co-inoculation of basal and SM cells was androgen responsive. In addition, a significant reduction in prostatic tissue was noted in animals treated with doxazosin. CONCLUSION: We have established an in vivo model that uses prostatic epithelial and SM cell lines for investigating cellular interactions between epithelial and SM cells that regulate prostatic growth and function. This model will be useful for delineating the mechanisms by which prostatic cells interact and in determining the efficacy of new approaches aimed at interfering with prostatic stromal/epithelial interactions that result in abnormal cellular proliferation.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Cell Communication , Doxazosin/pharmacology , Prostatic Hyperplasia/physiopathology , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/physiology , Immunohistochemistry , Male , Mice , Muscle, Smooth/cytology , Prostatic Hyperplasia/drug therapy , Stromal Cells/physiology
15.
Blood ; 100(10): 3527-35, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12411316

ABSTRACT

Recent experiments show that hematopoietic progenitor cell populations contain endothelial precursor cells. We have isolated a population of CD34(+) cells that expresses fibroblast growth factor receptor-1 (FGFR-1) and that differentiates into endothelial cells in vitro. We find that 4.5% +/- 2.1% of CD34(+) cells isolated from bone marrow, cord blood, and mobilized peripheral blood express FGFR-1 and that viable CD34(+)FGFR(+) cells are small, with little granularity, and express both primitive hematopoietic and endothelial markers on their surface. The primitive hematopoietic markers AC133, c-kit, and Thy-1 are coexpressed by 75%, 85%, and 64% of CD34(+)FGFR(+) cells, respectively. Most of the CD34(+)FGFR(+) cells also express antigens found on endothelial cells, such as CD31, vascular endothelial growth factor receptor-2, and the endothelial-specific cell surface marker, vascular endothelial cadherin (VE-cadherin), whereas 56% to 60% of the cells express Tie, Tek, and the endothelial-specific marker, P1H12. The CD34(+)FGFR(+) population is enriched in cells expressing endothelial-specific antigens compared with the CD34(+) population. Isolated CD34(+)FGFR(+) cells grow slowly in culture, are stimulated by fibroblast growth factor-2 and vascular endothelial growth factor, and give rise to cells that express von Willebrand factor and VE-cadherin and that incorporate acetylated low-density lipoprotein. These experiments show that FGFR-1 is expressed by a subpopulation of CD34(+) cells that give rise to endothelial cells in vitro, indicating that this population contains endothelial stem/progenitor cells.


Subject(s)
Endothelium/cytology , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Fibroblast Growth Factor/biosynthesis , Stem Cells/metabolism , Cell Culture Techniques , Cell Division , Cell Separation , Endothelial Growth Factors/pharmacology , Endothelium/immunology , Fibroblast Growth Factor 2/pharmacology , Flow Cytometry , Humans , Immunophenotyping , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphokines/pharmacology , Receptor Protein-Tyrosine Kinases/analysis , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/analysis , Stem Cells/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
J Cell Biol ; 157(7): 1257-65, 2002 Jun 24.
Article in English | MEDLINE | ID: mdl-12082083

ABSTRACT

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.


Subject(s)
Homeostasis , Prostate/cytology , Stem Cells/cytology , Animals , Cell Cycle , Cell Division , Cells, Cultured , Collagen/metabolism , Culture Media , Gels , Kinetics , Male , Mice , Mice, Inbred C57BL , Models, Biological , Prostate/anatomy & histology
17.
Prostate ; 51(3): 175-88, 2002 May 15.
Article in English | MEDLINE | ID: mdl-11967952

ABSTRACT

BACKGROUND: We have derived a panel of p53-null prostatic "basal" and "luminal" epithelial cell lines and their ras transformed counterparts to study stromal/epithelial interactions and the properties of tumors arising from "basal" and "luminal" cells. METHODS: Previously derived normal murine prostatic "basal" epithelial (PE-B-1) and "luminal" epithelial (PE-L-1) cell lines were transformed with N-Ras. These lines and a spontaneously transformed "luminal" cell line were inoculated subcutaneously or orthotopically into athymic mice, alone or in combination with normal prostatic smooth muscle cells (SMC). RESULTS: All transformed lines formed subcutaneous tumors. SMC significantly enhanced the growth rate of the tumors arising from the "basal" and one of the "luminal" cell lines. The transformed "basal" line gave rise to tumors expressing both "basal" and "luminal" cytokeratins. CONCLUSIONS: Prostatic SMC promote the growth of transformed epithelial cells, suggesting that prostatic stroma may promote tumor development. Furthermore, transformed "basal" cells give rise to tumors containing "luminal" cells, suggesting that although most human tumors have a "luminal" phenotype, they may originate from transformed "basal" cells.


Subject(s)
Prostate/physiology , Prostatic Neoplasms/pathology , Stromal Cells/physiology , Agar , Animals , Cell Differentiation/physiology , Cell Division/physiology , Cell Line, Transformed , Cell Transplantation , Culture Media , Dihydrotestosterone/pharmacology , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth/cytology , Neoplasm Transplantation , Phenotype , Prostate/cytology , Prostatic Neoplasms/genetics , Stromal Cells/transplantation , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology , Tumor Cells, Cultured
18.
J Cell Physiol ; 190(1): 83-91, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11807814

ABSTRACT

Basic fibroblast growth factor (bFGF) is produced by bone marrow stromal cells as well as by normal and leukemic hematopoietic cells. In this study, we examine the direct effects of bFGF on erythroid differentiation in K562 cells in order to determine whether bFGF can promote the expression of a primitive phenotype. Low levels of bFGF inhibited erythroid differentiation as evidenced by decreased expression of glycophorin A and increased expression of c-kit. bFGF also increased both the numbers and the sizes of colonies of K562 cells in soft agar assays. The addition of TGF-beta to these cells induced erythroid differentiation which resulted in an increase in glycophorin A and a decrease in c-kit. The simultaneous addition of bFGF and TGF-beta to K562 cells prevented both the TGF-beta-mediated increase in glycophorin A expression and the decrease in c-kit expression associated with erythroid differentiation. bFGF antagonised the TGF-beta-mediated promotion of erythroid differentiation in K562 cells in a dose dependent manner and these two cytokines counteracted each other on an approximately molar basis. These results indicate that bFGF alone increases expression of c-kit and promotes a primitive phenotype in K562 cells. In addition, bFGF counteracts the effects of differentiation-inducing cytokines, such as TGF-beta, on hematopoietic cells. It is therefore possible that enhanced production of bFGF by leukemic cells could contribute to their neoplastic phenotype by opposing the effects of negative regulators or cytokines that induce differentiation.


Subject(s)
Cell Differentiation/physiology , Fibroblast Growth Factor 2/metabolism , Glycophorins/metabolism , K562 Cells/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Cell Count , Cell Differentiation/drug effects , Erythrocytes/cytology , Erythrocytes/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glycophorins/genetics , Humans , K562 Cells/cytology , K562 Cells/drug effects , Microscopy, Fluorescence , Proto-Oncogene Proteins c-kit/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology
19.
Prostate ; 50(2): 83-91, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11816016

ABSTRACT

BACKGROUND: The vasculature of the prostate responds to androgens. Androgens most likely affect the vasculature indirectly by modulating the expression of angiogenic factors in the cells of the prostate. Most studies to date have examined the production of angiogenic factors by the prostate luminal epithelium. Here we examine the effects of androgen on production of three angiogenic factors, vascular endothelial growth factor (VEGF), angiopoietin-1, and angiopoietin-2, by the three major cell types in the prostate. METHODS: The ability of androgen to modulate VEGF, angiopoietin-1, and angiopoietin-2 production in cultured mouse prostate luminal epithelial, basal epithelial, and smooth muscle cells (SMCs) was assessed by Western blot and RT-PCR. RESULTS: The production of VEGF was modulated by androgens in both luminal epithelial and prostate SMCs but not in basal epithelial cells. However, in prostate luminal epithelial cell cultures, VEGF was predominately secreted apically, suggesting that in vivo most of the epithelium-derived VEGF is unavailable to the underlying blood vessels. In addition, prostate luminal epithelial cells produced angiopoietin-2, an angiogenesis inhibitor. In contrast, prostate SMCs produced angiopoietin-1, a positive modulator of angiogenesis. Synthesis of the angiopoietins did not respond to androgen treatment. CONCLUSIONS: Prostate smooth muscle may play an important role in regulating vascular responses to androgen.


Subject(s)
Androgens/pharmacology , Endothelial Growth Factors/biosynthesis , Gene Expression Regulation , Lymphokines/biosynthesis , Membrane Glycoproteins/biosynthesis , Prostate/physiology , Protein Biosynthesis , Angiopoietin-1 , Angiopoietin-2 , Animals , Cell Culture Techniques , Epithelial Cells/physiology , Male , Mice , Muscle, Smooth/physiology , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...