201
|
Hackett TL, Knight DA. The role of epithelial injury and repair in the origins of asthma. Curr Opin Allergy Clin Immunol 2007; 7:63-8. [PMID: 17218813 DOI: 10.1097/aci.0b013e328013d61b] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW We currently understand little about the mechanisms that lead to asthma. The bronchial epithelium is the first cell layer of contact with the environment and as such is an especially attractive target in which to identify novel mechanisms and new therapeutic strategies in disease development. We discuss the role of epithelial injury and wound repair in the origins of asthma. RECENT FINDINGS The presence of inflammation, thickening of the basement membrane and angiogenesis have been described in bronchial biopsies from asthmatic children. We and others have demonstrated the utility of bronchial brushings from children for the isolation, characterization and culture of primary epithelial cells. The results of these experiments suggest that intrinsic differences exist between asthmatic and nonasthmatic epithelial cells. SUMMARY It is becoming increasingly clear from studies involving adults and, more recently, children, that the epithelium orchestrates inflammatory and remodeling responses of the airway. Equally clear is that the asthmatic epithelium responds inappropriately to challenge and displays signs of dysregulated repair. Understanding the regulatory mechanisms involved in these processes, including the role of resident/recruited progenitor cells, is crucial if we are to halt the progression of asthma when the disease first manifests in childhood.
Collapse
Affiliation(s)
- Tillie-Louise Hackett
- James Hogg iCAPTURE Centre for Cardiovascular and Pulmonary Research, St Paul's Hospital, Vancouver, BC, Canada
| | | |
Collapse
|
202
|
Abstract
Identification of cancer stem cells (CSCs) in both hematological and solid malignancies suggests that CSCs may be a common phenomenon for most malignancies. Similarly to normal stem cells, CSCs can self-renew and differentiate into progeny cancer cells. Almost all current therapy against cancer targets differentiated cancer cells. CSCs are more resistant to therapy secondary to quiescence, increased expression of antiapoptotic proteins and drug efflux transporters. In this article, we review the current status of CSC research and propose the targeting of CSC cell-surface molecules, signal transduction pathways, the stem cell niche, stem cell differentiation and drug resistance.
Collapse
Affiliation(s)
- Chong-Xian Pan
- University of California at Davis Cancer Center, 4501 X Street, Room 3016, Sacramento, CA 95817, USA.
| | | | | |
Collapse
|
203
|
Tang DG, Patrawala L, Calhoun T, Bhatia B, Choy G, Schneider-Broussard R, Jeter C. Prostate cancer stem/progenitor cells: identification, characterization, and implications. Mol Carcinog 2007; 46:1-14. [PMID: 16921491 DOI: 10.1002/mc.20255] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Several solid tumors have now been shown to contain stem cell-like cells called cancer stem cells (CSC). These cells, although generally rare, appear to be highly tumorigenic and may be the cells that drive tumor formation, maintain tumor homeostasis, and mediate tumor metastasis. In this Perspective, we first provide our insight on how a CSC should be defined. We then summarize our current knowledge of stem/progenitor cells in the normal human prostate (NHP), an organ highly susceptible to hyperproliferative diseases such as benign prostate hyperplasia (BPH) and prostate cancer (PCa). We further review the evidence that cultured PCa cells, xenograft prostate tumors, and patient tumors may contain stem/progenitor cells. Along with our discussion, we present several methodologies that can be potentially used to identify putative tumor-reinitiating CSC. Finally, we present a hypothetical model for the hierarchical organization of human PCa cells and discuss the implications of this model in helping understand prostate carcinogenesis and design novel diagnostic, prognostic, and therapeutic approaches.
Collapse
Affiliation(s)
- Dean G Tang
- Department of Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park-Research Division, Smithville, Texas 78957, USA
| | | | | | | | | | | | | |
Collapse
|
204
|
Lieu DK, Degraffenried LA, Isseroff RR, Kurzrock EA. β1 Integrin Expression Pattern in Transitional Urothelium Does Not Allow for Efficient Stem Cell Enrichment as in Other Epithelia. ACTA ACUST UNITED AC 2007; 13:263-70. [PMID: 17518562 DOI: 10.1089/ten.2006.0132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
With a lack of distinct stem cell markers, isolation of tissue-specific stem cells for tissue engineering and gene therapy is a great challenge. Beta (beta)(1) integrin expression has been used as a way of enriching for putative epithelial stem cells through rapid adhesion to collagen IV or flow cytometry. This is a first report of enrichment of putative urothelial stem cells using rapid adhesion and flow cytometric methods. We assessed our success by determining the clonogenic and proliferative potential of the isolated cells. We demonstrated that enrichment based on beta(1) integrin expression with flow cytometry yields highly clonogenic and proliferative urothelial cells, whereas the rapid adhesion method is not as efficient, possibly because of the unique nature of urothelium, a transitional epithelium, compared to results reported in stratified and columnar epithelia.
Collapse
Affiliation(s)
- Deborah K Lieu
- Departments of Urology, University of California at Davis, Sacramento, California 95817, USA
| | | | | | | |
Collapse
|
205
|
Lang SH, Smith J, Hyde C, Macintosh C, Stower M, Maitland NJ. Differentiation of prostate epithelial cell cultures by matrigel/ stromal cell glandular reconstruction. In Vitro Cell Dev Biol Anim 2007; 42:273-80. [PMID: 17163777 DOI: 10.1290/0511080.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Accepted: 04/09/2006] [Indexed: 12/18/2022]
Abstract
Three-dimensional epithelial culture models are widely used to emulate a more physiologically relevant microenvironment for the study of genes and signaling pathways. Prostate epithelial cells can grow into solid cell masses or acinus-like spheroids in Matrigel. To test if the ability to form acinus-like spheroids in Matrigel is dependent on how undifferentiated a cell is or whether it is tumor or nontumor, we established six novel epithelial cell lines. Primary prostate epithelial cells were immortalized using HPV16 E6 gene transduction and were named Shmac 2, 3, and 6 (nontumor); Shmac 4, Shmac 5, and P4E6 (tumor). All cell lines were phenotyped in monolayer culture, and their ability to form acinus-like spheroids in Matrigel investigated. The cell lines exhibited a wide range of population doubling times and all showed an intermediate phenotype in monolayer culture ((luminal)CK(+)/(basal)CK(+)/CD44(+)/PSA(+)/AR(-)). Only Shmac 5 cells formed acinus-like spheroids when cultured in Matrigel. Co-culture of the spheroids with fibroblasts advanced differentiation by inducing androgen receptor expression and epithelial polarization. Our findings indicate that tumor cells can form acinus-like spheroids in Matrigel.
Collapse
Affiliation(s)
- Shona H Lang
- YCR Cancer Research Unit, University of York, UK.
| | | | | | | | | | | |
Collapse
|
206
|
Wang H, McKnight NC, Zhang T, Lu ML, Balk SP, Yuan X. SOX9 Is Expressed in Normal Prostate Basal Cells and Regulates Androgen Receptor Expression in Prostate Cancer Cells. Cancer Res 2007; 67:528-36. [PMID: 17234760 DOI: 10.1158/0008-5472.can-06-1672] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
SOX9 is a member of the SOX [Sry-related high-mobility group (HMG) box] family of HMG DNA-binding domain transcription factors and is required for the development and differentiation of multiple cell lineages. This report shows that basal epithelial cells express SOX9 in normal prostate, with no detectable expression in luminal epithelial cells. In contrast, SOX9 is expressed in primary prostate cancers in vivo, at a higher frequency in recurrent prostate cancer and in prostate cancer cell lines (LNCaP, CWR22, PC3, and DU145). SOX9 message and protein levels in prostate cancer cells were increased by treatment with glycogen synthase kinase 3beta inhibitor (SB415286), and SOX9 was reduced when beta-catenin was down-regulated by small interfering RNA (siRNA), indicating that SOX9 expression in prostate cancer is regulated by Wnt/beta-catenin signaling. SOX9 bound specifically to androgen receptor (AR) DNA-binding domain glutathione S-transferase fusion proteins, and this interaction was dependent on a short peptide immediately COOH-terminal to the DNA-binding domain (the C-terminal extension), which is required for interactions between steroid hormone receptors and the architectural HMG proteins. Exogenous SOX9 expressed at high nonphysiologic levels decreased AR expression and activity; however, at lower levels, SOX9 increased AR protein expression. Significantly, down-regulation of SOX9 by siRNA in prostate cancer cells reduced endogenous AR protein levels, and cell growth indicating that SOX9 contributes to AR regulation and decreased cellular proliferation. These results indicate that SOX9 in prostate basal cells supports the development and maintenance of the luminal epithelium and that a subset of prostate cancer cells may escape basal cell requirements through SOX9 expression.
Collapse
Affiliation(s)
- Hongyun Wang
- Cancer Biology Program, Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Urology Research Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | |
Collapse
|
207
|
Lawson DA, Xin L, Lukacs RU, Cheng D, Witte ON. Isolation and functional characterization of murine prostate stem cells. Proc Natl Acad Sci U S A 2006; 104:181-6. [PMID: 17185413 PMCID: PMC1716155 DOI: 10.1073/pnas.0609684104] [Citation(s) in RCA: 305] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The ability to isolate prostate stem cells is essential to explore their role in prostate development and disease. In vitro prostate colony- and sphere-forming assays were used to quantitatively measure murine prostate stem/progenitor cell enrichment and self-renewal. Cell surface markers were screened for their ability to positively or negatively enrich for cells with enhanced growth potential in these assays. Immunohistochemical and FACS analyses demonstrate that specific cell surface markers can be used to discriminate prostate stromal (CD34(+)), luminal epithelial (CD24(+)CD49f(-)), basal epithelial (CD24(+)CD49f(+)), hematopoietic (CD45(+), Ter119(+)), and endothelial (CD31(+)) lineages. Sorting for cells with a CD45(-)CD31(-)Ter119(-)Sca-1(+)CD49f(+) antigenic profile results in a 60-fold enrichment for colony- and sphere-forming cells. These cells can self-renew and expand to form spheres for many generations and can differentiate to produce prostatic tubule structures containing both basal and luminal cells in vivo. These cells also localize to the basal cell layer within the region of the gland that is proximal to the urethra, which has been identified as the prostate stem cell niche. Prostate stem cells can be isolated to a purity of up to 1 in 35 by using this antigenic profile. The remarkable similarity in cell surface profile between prostate and mammary gland stem cells suggests these markers may be conserved among epithelial stem cell populations.
Collapse
Affiliation(s)
- Devon A. Lawson
- *Departments of Microbiology, Immunology, and Molecular Genetics and
| | - Li Xin
- *Departments of Microbiology, Immunology, and Molecular Genetics and
| | - Rita U. Lukacs
- *Departments of Microbiology, Immunology, and Molecular Genetics and
| | - Donghui Cheng
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095
| | - Owen N. Witte
- *Departments of Microbiology, Immunology, and Molecular Genetics and
- Molecular and Medical Pharmacology, David Geffen School of Medicine, and
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
208
|
Abstract
The vitamin D receptor (VDR) is a member of the large family of nuclear receptor transcription factors and specifically binds the micronutrient-derived hormone 1alpha,25(OH)2D3. A central endocrine role for this receptor in bone health was established at the beginning of the 20th century. Over the last 25 years, additional roles, perhaps through autocrine and paracrine mechanisms, have been established for VDR to regulate cell proliferation and differentiation, and more recently to exert immunomodulatory and antimicrobial functions. These findings, from in vitro and in vivo experiments, have generated considerable interest in targeting the VDR in multiple therapeutic settings. As with many potential therapeutics, it has also become clear that cells and tissues may also display de novo and acquired mechanisms of resistance to these actions. Consequently, a range of experimental and clinical options are being developed to bring about more targeted actions, overcome resistance and enhance efficacy of VDR-centred therapeutics.
Collapse
Affiliation(s)
- Moray J Campbell
- Institute of Biomedical Research, Wolfson Drive, University of Birmingham Medical School, Edgbaston, Birmingham, B15 2TT, UK.
| | | |
Collapse
|
209
|
Kasper S, Cookson MS. Mechanisms leading to the development of hormone-resistant prostate cancer. Urol Clin North Am 2006; 33:201-10, vii. [PMID: 16631458 DOI: 10.1016/j.ucl.2005.12.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Advanced and metastatic prostate cancers remain potentially lethal tumors. Although androgen deprivation therapy remains the most effective treatment, patients who progress to androgen independence die of their disease. This article focuses on the mechanisms by which hormone resistance develops, including the reactivation of androgen receptor during androgen deprivation therapy, the role of cancer stem cells, and the emergence of epithelial-mesenchymal transition cells, which have increased metastatic potential. It is through an enhanced understanding of these mechanisms that new therapies can be developed to combat this disease.
Collapse
Affiliation(s)
- Susan Kasper
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232-2765, USA.
| | | |
Collapse
|
210
|
Abstract
Prostate cancer is the most frequently diagnosed cancer in men. Despite recent advances in the detection of early prostate cancer there is little effective therapy for patients with locally advanced and/or metastatic disease. The majority of patients with advanced disease respond initially to androgen ablation therapy. However, most go on to develop androgen-independent tumours that inevitably are fatal. A similar response is seen to chemotherapeutic and radiotherapy treatments. As a result, metastatic prostate cancer remains an incurable disease by current treatment strategies. Recent reports of cancer stem cells have prompted questions regarding the involvement of normal stem/progenitor cells in prostate tumour biology, their potential contribution to the tumour itself and whether they are the cause of tumour initiation and progression. Although still controversial, the cancer stem cell is likely to be the most crucial target in the treatment of prostate cancer, and a thorough understanding of its biology, particularly of how the cancer stem cell differs from the normal stem cell, might allow it to be targeted selectively and eliminated, thus improving therapeutic outcome.
Collapse
Affiliation(s)
- Anne T Collins
- University of York, YCR Cancer Research Unit, Department of Biology, Heslington, York YO10 5DD, UK.
| | | |
Collapse
|
211
|
Goto K, Salm SN, Coetzee S, Xiong X, Burger PE, Shapiro E, Lepor H, Moscatelli D, Wilson EL. Proximal prostatic stem cells are programmed to regenerate a proximal-distal ductal axis. Stem Cells 2006; 24:1859-68. [PMID: 16644920 DOI: 10.1634/stemcells.2005-0585] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
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.
Collapse
Affiliation(s)
- Ken Goto
- Department of Cell Biology, MSB 634, NYU School of Medicine, 550 First Avenue, New York, New York 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Heer R, Collins AT, Robson CN, Shenton BK, Leung HY. KGF suppresses α2β1 integrin function and promotes differentiation of the transient amplifying population in human prostatic epithelium. J Cell Sci 2006; 119:1416-24. [PMID: 16554439 DOI: 10.1242/jcs.02802] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Prostate epithelial stem cells are self-renewing cells capable of differentiation into prostate epithelium, and are thought to contribute towards both benign and malignant conditions in the human prostate. We have previously demonstrated that prostate epithelial basal cells express high levels of integrin α2β1 and this population can be subdivided into stem (α2β1hi CD133+) and transient-amplifying population (TAP) cells (α2β1hi CD133-). However, the molecular mechanism(s) controlling the commitment and regulation of these cells towards differentiated epithelium remains unclear. Here, we demonstrate that β1 integrin function is required for the maintenance of basal prostatic epithelial cells and suppression of its function by either methylcellulose or, more specifically, β1-blocking antibody (80 μg/ml) induces differentiation, with associated expression of the differentiation-specific markers prostate acid phosphatase (PAP) and cytokeratin 18 (CK18). Keratinocyte growth factor (KGF), a stromal-derived growth factor, has previously been implicated in prostate organogenesis using in vitro tissue recombination experiments. We show that treatment with KGF (10 ng/ml) potently induces epithelial differentiation with concomitant suppression of α2β1 integrin expression as well as the induction of androgen receptor expression. Specifically, p38-MAPK appears to be involved and the presence of SB202190, a p38 inhibitor, significantly blocks KGF-induced differentiation. Furthermore, the expression of the high-affinity receptor tyrosine kinase to KGF (FGFR2) is predominantly detectable in α2β1hi CD133- TAP cells when compared with stem cells (α2β1hi CD133+), which would therefore be relatively unresponsive to the differentiating effect of KGF. Taken together, using a human primary culture model, we have demonstrated key roles for interactions between KGF and integrin-mediated function in the regulation of prostate epithelial differentiation.
Collapse
Affiliation(s)
- Rakesh Heer
- Urology Research Group, Northern Institute for Cancer Research, University of Newcastle, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| | | | | | | | | |
Collapse
|
213
|
Badia E, Briz MD, Pinart E, Sancho S, Garcia-Gil N, Bassols J, Pruneda A, Bussalleu E, Yeste M, Casas I, Bonet S. Structural and ultrastructural features of boar seminal vesicles. Tissue Cell 2006; 38:79-91. [PMID: 16533514 DOI: 10.1016/j.tice.2005.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Revised: 12/01/2005] [Accepted: 12/13/2005] [Indexed: 01/23/2023]
Abstract
The morphological features of boar seminal vesicles were examined by light and transmission microscopy. Boar seminal vesicles consist of glandular tissue arranged in multiple lobules containing a system of ramified secretory tubules. The secretory tubules are composed of a mucosa formed by an epithelium and an underlying lamina propria and, are surrounded by a muscular layer. The epithelium is made up of columnar cells and occasional basal cells. Mast cells are frequently found among epithelial cells. Three types of columnar cells, considered different stages of the secretory cell cycle, are present: principal cells, clear cells and dense cells. Principal cells are functionally differentiated cells characterised by abundant mitochondria, great development of the rough endoplasmic reticulum and presence of secretory granules in their cytoplasm. The apical surface of many principal cells shows apical blebs filled with PAS-positive material. No acid mucosubstances are detected. Microvilli cover the apical surface except in the apical blebs. Dense cells, arranged between principal cells, are also functional differentiated cells but with signs of cellular degeneration. Clear cells are an initial differentiated stage of columnar cells and are characterised by the presence of a poorly developed rough endoplasmic reticulum and by the absence of secretory granules. Proliferating cells are present among columnar cells. Basal cells contain scarce cytoplasm, few organelles and no secretory granules. The lack of mitotic activity in these cells suggests that they do not act as precursors of columnar cells.
Collapse
Affiliation(s)
- E Badia
- Biotechnology of Porcine Reproduction, Institute of Agroalimentary Technology (INTEA), Faculty of Sciences, University of Girona, Campus de Montilivi 17071, Girona, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Gustafson MP, Xu C, Grim JE, Clurman BE, Knudsen BS. Regulation of cell proliferation in a stratified culture system of epithelial cells from prostate tissue. Cell Tissue Res 2006; 325:263-76. [PMID: 16557385 DOI: 10.1007/s00441-005-0093-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Accepted: 09/26/2005] [Indexed: 01/01/2023]
Abstract
Mechanisms controlling epithelial proliferation and differentiation in the prostate have been primarily investigated in mouse models. The regulation of proliferation and differentiation is poorly understood in human prostate epithelial cells. In vivo, the glandular prostate epithelium consists of a p63-positive proliferating basal cell layer and a post-mitotic p27-positive secretory cell layer. We have established an organized stratified culture system of human primary prostate epithelial cells to gain insight into mechanisms regulating proliferation and differentiation. In this system, expression of p63 is observed in the bottom layer. In addition, BrdU incorporation persists even though cells are confluent. In contrast, in the upper layer, p63 expression is greatly diminished, p27 is expressed, and the cells are growth arrested. Overexpression of cyclin D1 or knockdown of p27 does not increase proliferation. After inactivation of the nuclear phosphoprotein Rb, the cell layers remain organized and cell proliferation increases only in the bottom layer. Furthermore, the expression of p63 remains confined to the bottom layer after Rb inactivation. Altogether, this in vitro model recapitulates certain aspects of in vivo growth regulation and differentiation and suggests that the loss of Rb family proteins in human cells trigger hyperplasia but is not sufficient for transformation.
Collapse
Affiliation(s)
- Michael P Gustafson
- Clinical Research and Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | |
Collapse
|
215
|
Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, Tang S, Reilly JG, Chandra D, Zhou J, Claypool K, Coghlan L, Tang DG. Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene 2006; 25:1696-708. [PMID: 16449977 DOI: 10.1038/sj.onc.1209327] [Citation(s) in RCA: 715] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD44 is a multifunctional protein involved in cell adhesion and signaling. The role of CD44 in prostate cancer (PCa) development and progression is controversial with studies showing both tumor-promoting and tumor-inhibiting effects. Most of these studies have used bulk-cultured PCa cells or PCa tissues to carry out correlative or overexpression experiments. The key experiment using prospectively purified cells has not been carried out. Here we use FACS to obtain homogeneous CD44(+) and CD44(-) tumor cell populations from multiple PCa cell cultures as well as four xenograft tumors to compare their in vitro and in vivo tumor-associated properties. Our results reveal that the CD44(+) PCa cells are more proliferative, clonogenic, tumorigenic, and metastatic than the isogenic CD44(-) PCa cells. Subsequent molecular studies demonstrate that the CD44(+) PCa cells possess certain intrinsic properties of progenitor cells. First, BrdU pulse-chase experiments reveal that CD44(+) cells colocalize with a population of intermediate label-retaining cells. Second, CD44(+) PCa cells express higher mRNA levels of several 'stemness' genes including Oct-3/4, Bmi, beta-catenin, and SMO. Third, CD44(+) PCa cells can generate CD44(-) cells in vitro and in vivo. Fourth, CD44(+) PCa cells, which are AR(-), can differentiate into AR(+) tumor cells. Finally, a very small percentage of CD44(+) PCa cells appear to undergo asymmetric cell division in clonal analyses. Altogether, our results suggest that the CD44(+) PCa cell population is enriched in tumorigenic and metastatic progenitor cells.
Collapse
Affiliation(s)
- L Patrawala
- Department of Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park-Research Division, Smithville, 78957, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 2006; 65:10946-51. [PMID: 16322242 DOI: 10.1158/0008-5472.can-05-2018] [Citation(s) in RCA: 2030] [Impact Index Per Article: 106.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Existing therapies for prostate cancer eradicates the bulk of cells within a tumor. However, most patients go on to develop androgen-independent disease that remains incurable by current treatment strategies. There is now increasing evidence in some malignancies that the tumor cells are organized as a hierarchy originating from rare stem cells that are responsible for maintaining the tumor. We report here the identification and characterization of a cancer stem cell population from human prostate tumors, which possess a significant capacity for self-renewal. These cells are also able to regenerate the phenotypically mixed populations of nonclonogenic cells, which express differentiated cell products, such as androgen receptor and prostatic acid phosphatase. The cancer stem cells have a CD44+/alpha2beta1hi/CD133+ phenotype, and we have exploited these markers to isolate cells from a series of prostate tumors with differing Gleason grade and metastatic states. Approximately 0.1% of cells in any tumor expressed this phenotype, and there was no correlation between the number of CD44+/alpha2beta1hi/CD133+ cells and tumor grade. The identification of a prostate cancer stem cell provides a powerful tool to investigate the tumorigenic process and to develop therapies targeted to the stem cell.
Collapse
Affiliation(s)
- Anne T Collins
- Yorkshire Cancer Research Unit, Department of Biology, University of York, York, United Kingdom.
| | | | | | | | | |
Collapse
|
217
|
Abstract
The prostate gland is the site of the most commonly diagnosed cancer in men in USA and UK, accounting for one in five of new cases of male cancer. Common with many other cancer types, prostate cancer is believed to arise from a stem cell that shares characteristics with the normal stem cell. Normal prostate epithelial stem cells were recently identified and found to have a basal cell phenotype together with expression of CD133. Preliminary data have now emerged for a prostate cancer stem cell that also expresses cell surface CD133 but lacks expression of the androgen receptor. Here we examine the evidence supporting the existence of prostate cancer stem cells and discuss possible mechanisms of stem cell maintenance.
Collapse
Affiliation(s)
- S Rizzo
- The Bob Champion Prostate Stem Cell Laboratory, Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, UK, SM2 5NG
| | | | | |
Collapse
|
218
|
Maitland NJ, Bryce SD, Stower MJ, Collins AT. Prostate cancer stem cells: a target for new therapies. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2006:155-179. [PMID: 17939301 DOI: 10.1007/2789_2007_050] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Prostate cancer is now a common disease in men over 50 years of age. Medical therapies for prostate cancer are based on discoveries from the mid-twentieth century, and in the long term are rarely curative. Most treatments are directed towards an androgen receptor-expressing, highly proliferative target cell, which does indeed form the vast majority of cells in a prostate tumour. However, by invoking the existence of a cancer stem cell which, like normal epithelial stem cells in the prostate, does not express androgen receptor and is relatively quiescent, the observed resistance to most medical therapies can be explained. The phenotype of the prostate cancer stem cells is that of a basal cell and cultures derived from cancers, but not benign tissues, express a range of prostate cancer-associated RNAs. Furthermore, stem cells purified on the basis of alpha2beta1 high integrin and CD133 cell surface antigen expression, from an established culture of Gleason 4 (2+2) prostate cancer (P4E6), were able to form multiple intraprostatic tumours in nude mice when grafted orthotopically in a matrigel plug containing human prostatic stroma. The final tumours reexpressed androgen receptor and displayed a histology similar to that of a Gleason 4 cancer.
Collapse
Affiliation(s)
- N J Maitland
- Department of Biology, YCR Cancer Research Unit, University of York, YO10 5YW York, UK.
| | | | | | | |
Collapse
|
219
|
Campos LS, Decker L, Taylor V, Skarnes W. Notch, epidermal growth factor receptor, and beta1-integrin pathways are coordinated in neural stem cells. J Biol Chem 2005; 281:5300-9. [PMID: 16332675 DOI: 10.1074/jbc.m511886200] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Notch1 and beta1-integrins are cell surface receptors involved in the recognition of the niche that surrounds stem cells through cell-cell and cell-extracellular matrix interactions, respectively. Notch1 is also involved in the control of cell fate choices in the developing central nervous system (Lewis, J. (1998) Semin. Cell Dev. Biol. 9, 583-589). Here we report that Notch and beta1-integrins are co-expressed and that these proteins cooperate with the epidermal growth factor receptor in neural progenitors. We describe data that suggests that beta1-integrins may affect Notch signaling through 1) physical interaction (sequestration) of the Notch intracellular domain fragment by the cytoplasmic tail of the beta1-integrin and 2) affecting trafficking of the Notch intracellular domain via caveolin-mediated mechanisms. Our findings suggest that caveolin 1-containing lipid rafts play a role in the coordination and coupling of beta1-integrin, Notch1, and tyrosine kinase receptor signaling pathways. We speculate that this will require the presence of the adequate beta1-activating extracellular matrix or growth factors in restricted regions of the central nervous system and namely in neurogenic niches.
Collapse
Affiliation(s)
- Lia Scotti Campos
- INSERM U368, Biologie Moléculaire du Développement, Ecole Normale Supérieure, Paris, France.
| | | | | | | |
Collapse
|
220
|
Abstract
Cancer stem cells undoubtedly exist in many tumour types, including the prostate. This hypothesis can explain both the heterogeneity of prostate tumours and their variable responses to several conventional therapies. In the longer term, therapies directed against tumour stem cells should offer a real possibility of long-term cure, rather than current palliative therapy. Identifying specific tumour stem-cell markers will enhance this process, but the scarcity of these cells within the mass of more differentiated amplifying progeny that comprise >99.9% of most cancers makes this a severe technical challenge. In addition, many tumour stem-cell markers are probably shared with normal stem cells, both in prostate and in stem cells from other tissues, but tumour-specific patterns of gene expression, probably designed to allow the tumour stem cell to survive outside its protective 'niche' in normal tissues, will be the best initial targets for new therapeutic agents.
Collapse
Affiliation(s)
- Norman J Maitland
- YCR Cancer Research Unit, Department of Biology, University of York, York, UK.
| | | |
Collapse
|
221
|
Abstract
As prostate cancer is not a single disease, it is important to identify the pivotal pathway in the patient being treated. The molecular environment is the site of current oncological research to define new therapeutic targets for hormone-refractory disease, offering the potential to eventually individualize treatment through stratification of pathways. Targets may be validated either phenotypically (e.g. androgen receptor, cadherin) or functionally (e.g. prostate cancer-specific genes). In addition, several other candidates are potentially suitable, while others await discovery. Important initial steps have been made in the search for prostate cancer stem cells; identifying stem cells and the stromal, hormonal, and other signalling molecules that influence their behaviour would have important implications for managing prostate cancer. Although individual therapeutic pathways might be ineffective in a particular molecular environment, combinations of approaches might be capable of producing synergistic effects. A multimodal approach thus might be the best solution. Determining where best to search for a molecular target, and validating whether the target is associated with a sufficiently aggressive malignant process to justify further study is difficult, but the potential benefits are enormous.
Collapse
Affiliation(s)
- Jack A Schalken
- Department of Experimental Urology, Radboud University Nijmegen Medical Center, Geert Grooteplein 30, Nijmegen, the Netherlands.
| |
Collapse
|
222
|
Kanatsu-Shinohara M, Inoue K, Lee J, Miki H, Ogonuki N, Toyokuni S, Ogura A, Shinohara T. Anchorage-independent growth of mouse male germline stem cells in vitro. Biol Reprod 2005; 74:522-9. [PMID: 16306420 DOI: 10.1095/biolreprod.105.046441] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Spermatogenesis originates from a small number of spermatogonial stem cells that reside on the basement membrane and undergo self-renewal division to support spermatogenesis throughout the life of adult animals. Although the recent development of a technique to culture spermatogonial stem cells allowed reproduction of self-renewal division in vitro, much remains unknown about how spermatogonial stem cells are regulated. In this study, we found that spermatogonial stem cells could be cultured in an anchorage-independent manner, which is characteristic of stem cells from other types of self-renewing tissues. Although the cultured cells grew slowly (doubling time, approximately 4.7 days), they expressed markers of spermatogonia, and grew exponentially for at least 5 months to achieve 1.5 x 10(10) -fold expansion. The cultured cells underwent spermatogenesis following transplantation into the seminiferous tubules of infertile animals and fertile offspring were obtained by microinsemination of germ cells that had developed within the testes of recipients of the cultured cells. These results indicate that spermatogonial stem cells can undergo anchorage-independent, self-renewal division, and suggest that stem cells have the common property to survive and proliferate in the absence of exogenous substrata.
Collapse
Affiliation(s)
- Mito Kanatsu-Shinohara
- Horizontal Medical Research Organization, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
223
|
|
224
|
Azuma M, Hirao A, Takubo K, Hamaguchi I, Kitamura T, Suda T. A quantitative matrigel assay for assessing repopulating capacity of prostate stem cells. Biochem Biophys Res Commun 2005; 338:1164-70. [PMID: 16286093 DOI: 10.1016/j.bbrc.2005.10.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2005] [Accepted: 10/12/2005] [Indexed: 10/25/2022]
Abstract
Homeostasis of prostate tissue is maintained by stem cells, although such cells have not been well characterized. Here, we report establishment of such a method using matrigel. Matrigel containing a single-cell suspension from adult prostatic cells was subcutaneously grafted into the flank of nude mice. Prostatic duct-like structures derived from donor tissue were observed in the gel 2 weeks after transplantation. Luminal and basal cells observed in the gel expressed several markers characteristic of prostatic and/or epithelial cells. When a mixture with both EGFP-positive and negative prostate cells was transplanted, prostatic ducts consisted of either EGFP-positive or negative cells and chimeric patterns were rarely observed, suggesting that ducts were reconstituted from a single cell. Stem cell number and function were also evaluated by competition with control cells. Overall this method revealed that cells localized in the proximal portion in prostate ducts had higher reconstitution capacity than those in the distal portion. We conclude that prostate stem/progenitor cells exist and that our method is applicable to analysis of prostate stem cells, epithelial mesenchyme interactions, and prostate cancer stem cells.
Collapse
Affiliation(s)
- Masaki Azuma
- Department of Cell Differentiation, Keio University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
225
|
Leow CC, Wang XD, Gao WQ. Novel method of generating prostate-specific Cre-LoxP gene switching via intraductal delivery of adenovirus. Prostate 2005; 65:1-9. [PMID: 15791629 DOI: 10.1002/pros.20244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND In order to facilitate elucidation of oncogene or tumor suppressor gene function on initiation and progression of prostate cancer, it would be advantageous to develop an effective method to generate spatially and temporally controlled gene modification in murine prostates. METHODS Adenovirus expressing Cre-recombinase (Adeno-Cre) was intraductally injected into the prostate of ROSA26 reporter mice. Immmunohistochemical and X-gal staining were performed on prostate tissue sections harvested from mice at various time points following viral injection to confirm expression and activity of Cre-recombinase, respectively. RESULTS Adenovirus was intraductally delivered to the anterior lobe of the mouse prostate. Using this method of intraductal injection, we were able to precisely obtain Adeno-Cre infection to a majority of epithelial cells but not in the stromal cells or other organs. We further demonstrated that Adeno-Cre infected epithelial cells not only expressed Cre-recombinase enzyme but more importantly, Cre-recombinase activity was revealed through positive X-gal staining in Rosa26 reporter mice, thus, confirming epithelial-specific Cre-loxP recombination in Adeno-Cre infected prostate tissue sections. CONCLUSIONS This novel method of direct genetic delivery into adult murine prostates could provide an alternative to the more expensive and time-consuming transgenic/knockout approaches. The latter also have other limitations such as the availability of cell-type specific or temporally-regulated promoters, and the complication of genetic background differences, which can potentially be complemented by the technology we describe here.
Collapse
Affiliation(s)
- Ching Ching Leow
- Department of Molecular Biology, Genentech, Inc., South San Francisco, California 94080, USA
| | | | | |
Collapse
|
226
|
Riddick ACP, Shukla CJ, Pennington CJ, Bass R, Nuttall RK, Hogan A, Sethia KK, Ellis V, Collins AT, Maitland NJ, Ball RY, Edwards DR. Identification of degradome components associated with prostate cancer progression by expression analysis of human prostatic tissues. Br J Cancer 2005; 92:2171-80. [PMID: 15928670 PMCID: PMC2361819 DOI: 10.1038/sj.bjc.6602630] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Extracellular proteases of the matrix metalloproteinase (MMP) and serine protease families participate in many aspects of tumour growth and metastasis. Using quantitative real-time RT–PCR analysis, we have undertaken a comprehensive survey of the expression of these enzymes and of their natural inhibitors in 44 cases of human prostate cancer and 23 benign prostate specimens. We found increased expression of MMP10, 15, 24, 25 and 26, urokinase plasminogen activator-receptor (uPAR) and plasminogen activator inhibitor-1 (PAI1), and the newly characterised serine proteases hepsin and matriptase-1 (MTSP1) in malignant tissue compared to benign prostate tissue. In contrast, there was significantly decreased expression of MMP2 and MMP23, maspin, and the protease inhibitors tissue inhibitor of metalloproteinase 3 (TIMP3), TIMP4 and RECK (reversion-inducing cysteine-rich protein with Kazal motifs) in the cancer specimens. The expression of MMP15 and MMP26 correlated positively with Gleason score, whereas TIMP3, TIMP4 and RECK expression correlated negatively with Gleason score. The cellular localisation of the expression of the deregulated genes was evaluated using primary malignant epithelial and stromal cell cultures derived from radical prostatectomy specimens. MMP10 and 25, hepsin, MTSP1 and maspin showed predominantly epithelial expression, whereas TIMP 3 and 4, RECK, MMP2 and 23, uPAR and PAI1 were produced primarily by stromal cells. These data provide the first comprehensive and quantitative analysis of the expression and localisation of MMPs and their inhibitors in human prostate cancer, leading to the identification of several genes involved in proteolysis as potential prognostic indicators, in particular hepsin, MTSP1, MMP26, PAI1, uPAR, MMP15, TIMP3, TIMP4, maspin and RECK.
Collapse
Affiliation(s)
- A C P Riddick
- Norfolk and Norwich University Hospital NHS Trust, Norwich NR4 7UY, UK
| | - C J Shukla
- Norfolk and Norwich University Hospital NHS Trust, Norwich NR4 7UY, UK
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - C J Pennington
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - R Bass
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - R K Nuttall
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - A Hogan
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - K K Sethia
- Norfolk and Norwich University Hospital NHS Trust, Norwich NR4 7UY, UK
| | - V Ellis
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - A T Collins
- YCR Cancer Research Unit, Department of Biology, University of York, YO 10 5YW, UK
| | - N J Maitland
- YCR Cancer Research Unit, Department of Biology, University of York, YO 10 5YW, UK
| | - R Y Ball
- Norfolk and Norwich University Hospital NHS Trust, Norwich NR4 7UY, UK
| | - D R Edwards
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
- e-mail:
| |
Collapse
|
227
|
Schmelz M, Moll R, Hesse U, Prasad AR, Gandolfi JA, Hasan SR, Bartholdi M, Cress AE. Identification of a stem cell candidate in the normal human prostate gland. Eur J Cell Biol 2005; 84:341-54. [PMID: 15819412 PMCID: PMC2730953 DOI: 10.1016/j.ejcb.2004.12.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Stem cells of the human prostate gland have not yet been identified utilizing a structural biomarker. We have discovered a new prostatic epithelial cell phenotype-expressing cytokeratin 6a (Ck6a+ cells). The Ck6a+ cells are present within a specialized niche in the basal cell compartment in fetal, juvenile and adult prostate tissue, and within the stem cell-enriched urogenital sinus. In adult normal prostate tissue, the average abundance of Ck6a+ cells was 4.9%. With proliferative stimuli in the prostate organ culture model, in which the epithelial-stromal interaction was maintained, a remarkable increase of Ck6a expression was noticed to up to 64.9%. The difference in cytokeratin 6a expression between the normal adult prostate and the prostate organ culture model was statistically significant (p<0.0001). Within the prostate organ culture model the increase of cytokeratin 6a-expressing cells significantly correlated with increased proliferation index (r = 0.7616, p = 0.0467). The Ck6a+ cells were capable of differentiation as indicated by their expression of luminal cell markers such as ZO-1 and prostate specific antigen (PSA). Our data indicate that Ck6a+ cells represent a prostatic epithelial stem cell candidate possessing high potential for proliferation and differentiation. Since the development of benign prostatic hyperplasia and prostate carcinogenesis are disorders of proliferation and differentiation, the Ck6a+ cells may represent a major element in the development of these diseases.
Collapse
Affiliation(s)
- Monika Schmelz
- Department of Pathology, Southern Arizona Veterans Affairs Health Care System, 3601 S. 6th Ave., Tucson, AZ 85723, USA.
| | | | | | | | | | | | | | | |
Collapse
|
228
|
Burger PE, Xiong X, Coetzee S, Salm SN, Moscatelli D, Goto K, Wilson EL. Sca-1 expression identifies stem cells in the proximal region of prostatic ducts with high capacity to reconstitute prostatic tissue. Proc Natl Acad Sci U S A 2005; 102:7180-5. [PMID: 15899981 PMCID: PMC1129148 DOI: 10.1073/pnas.0502761102] [Citation(s) in RCA: 215] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
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.
Collapse
Affiliation(s)
- Patricia E Burger
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa.
| | | | | | | | | | | | | |
Collapse
|
229
|
Leone DP, Relvas JB, Campos LS, Hemmi S, Brakebusch C, Fässler R, Ffrench-Constant C, Suter U. Regulation of neural progenitor proliferation and survival by beta1 integrins. J Cell Sci 2005; 118:2589-99. [PMID: 15928047 DOI: 10.1242/jcs.02396] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neural stem cells give rise to undifferentiated nestin-positive progenitors that undergo extensive cell division before differentiating into neuronal and glial cells. The precise control of this process is likely to be, at least in part, controlled by instructive cues originating from the extracellular environment. Some of these cues are interpreted by the integrin family of extracellular matrix receptors. Using neurosphere cell cultures as a model system, we show that beta1-integrin signalling plays a crucial role in the regulation of progenitor cell proliferation, survival and migration. Following conditional genetic ablation of the beta1-integrin allele, and consequent loss of beta1-integrin cell surface protein, mutant nestin-positive progenitor cells proliferate less and die in higher numbers than their wild-type counterparts. Mutant progenitor cell migration on different ECM substrates is also impaired. These effects can be partially compensated by the addition of exogenous growth factors. Thus, beta1-integrin signalling and growth factor signalling tightly interact to control the number and migratory capacity of nestin-positive progenitor cells.
Collapse
Affiliation(s)
- Dino P Leone
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, ETH Hönggerberg, Zürich
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Li DQ, Chen Z, Song XJ, de Paiva CS, Kim HS, Pflugfelder SC. Partial enrichment of a population of human limbal epithelial cells with putative stem cell properties based on collagen type IV adhesiveness. Exp Eye Res 2005; 80:581-90. [PMID: 15781286 PMCID: PMC2906384 DOI: 10.1016/j.exer.2004.11.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Accepted: 11/24/2004] [Indexed: 12/30/2022]
Abstract
The concept that corneal epithelium stem cells reside in limbus has been recognized for more than a decade, but isolation of these stem cells has not been accomplished. This study was an initial attempt to isolate a population of human limbal epithelial cells enriched for certain putative stem cell properties based on their phenotype. Epithelial cells harvested from fresh human limbal rings and their primary cultures were allowed to adhere to collagen IV-coated dishes for 20 min and 2 hr, sequentially. The rapidly adherent cells (RAC), slowly adherent cells and non-adherent cells were evaluated for certain stem cell properties: (a) BrdU-label retention, (b) expression of basal cell (integrin beta1, p63, ABCG2) and differentiation (involucrin, keratin 12) markers, and (c) colony forming efficiency (CFE) and growth capacity on a 3T3 fibroblast feeder layer. Among unfractionated cells and the three selected populations, the RAC, accounting for about 10% of whole population, were enriched 5-fold in BrdU label-retaining cells, displayed the highest number of integrin beta1 and p63 positive and involucrin negative cells, expressed high levels of DeltaNp63 and ABCG2 mRNA, and lacked involucrin and K12 expression, and possessed the greatest CFE and growth capacity. These findings demonstrated for the first time that human limbal epithelial cells with stem cell properties can be partially enriched by their adhesiveness to collagen IV. The RAC population enriched for certain putative stem cell properties may prove useful in the future for transplantation to diseased and damaged corneas with limbal stem cell deficiency.
Collapse
Affiliation(s)
- De-Quan Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, 6565 Fannin Street, NC-205, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
231
|
Bhatia B, Tang S, Yang P, Doll A, Aumüeller G, Newman RA, Tang DG. Cell-autonomous induction of functional tumor suppressor 15-lipoxygenase 2 (15-LOX2) contributes to replicative senescence of human prostate progenitor cells. Oncogene 2005; 24:3583-95. [PMID: 15750631 DOI: 10.1038/sj.onc.1208406] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Normal human prostatic (NHP) epithelial cells undergo senescence in vitro and in vivo, but little is known about the tissue-specific molecular mechanisms. Here we first characterize young primary NHP cells as CK5(+)/CK18(+) intermediate basal cells that also express several other putative stem/progenitor cell markers including p63, CD44, alpha2beta1, and hTERT. When cultured in serum- and androgen-free medium, NHP cells gradually lose the expression of these markers, slow down in proliferation, and enter senescence. Several pieces of evidence implicate 15-lipoxygenase 2 (15-LOX2), a molecule with a restricted tissue expression and most abundantly expressed in adult human prostate, in the replicative senescence of NHP cells. First, the 15-LOX2 promoter activity and the mRNA and protein levels of 15-LOX2 and its multiple splice variants are upregulated in serially passaged NHP cells, which precede replicative senescence and occur in a cell-autonomous manner. Second, all immortalized prostate epithelial cells and prostate cancer cells do not express 15-LOX2. Third, PCa cells stably transfected with 15-LOX2 or 15-LOX2sv-b, a splice variant that does not possess arachidonate-metabolizing activity, show a passage-related senescence-like phenotype. Fourth, infection of early-passage NHP cells with retroviral vectors encoding 15-LOX2 or 15-LOX2sv-b induces partial cell-cycle arrest and big and flat senescence-like phenotype. Finally, 15-LOX2 protein expression in human prostate correlates with age. Together, these data suggest that 15-LOX2 may represent an endogenous prostate senescence gene and its tumor-suppressing functions might be associated with its ability to induce cell senescence.
Collapse
Affiliation(s)
- Bobby Bhatia
- Department of Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park-Research Division, 1808 Park Rd. 1C, Smithville, TX 78957, USA
| | | | | | | | | | | | | |
Collapse
|
232
|
Long RM, Morrissey C, Fitzpatrick JM, Watson RWG. Prostate epithelial cell differentiation and its relevance to the understanding of prostate cancer therapies. Clin Sci (Lond) 2005; 108:1-11. [PMID: 15384949 DOI: 10.1042/cs20040241] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Prostate cancer is the most common malignancy in males in the western world. However, little is known about its origin and development. This review highlights the biology of the normal prostate gland and the differentiation of basal epithelial cells to a secretory phenotype. Alterations in this differentiation process leading to cancer and androgen-independent disease are discussed, as well as a full characterization of prostate epithelial cells. A full understanding of the origin and characteristics of prostate cancer epithelial cells will be important if we are to develop therapeutic strategies to combat the heterogeneous nature of this disease.
Collapse
Affiliation(s)
- Ronan M Long
- Department of Surgery, Mater Misericordiae University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Republic of Ireland.
| | | | | | | |
Collapse
|
233
|
Lawson DA, Xin L, Lukacs R, Xu Q, Cheng D, Witte ON. Prostate stem cells and prostate cancer. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2005; 70:187-96. [PMID: 16869753 DOI: 10.1101/sqb.2005.70.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Understanding prostate stem cells (PSCs) may provide insight for the design of therapeutics for prostate cancer. We have developed a quantitative in vivo colony-forming assay and have demonstrated that the Sca-1 antigen is present on the surface of a prostate cell subpopulation that possesses multiple stem cell properties. Immunofluorescent analysis demonstrates that Sca-1 is expressed by both basal and luminal cells in the proximal region of the adult prostate, but is not expressed by either lineage in more distal regions. The proximal region has been suggested as the PSC niche based on BrdU label-retention studies and the presence of distinct smooth-muscle cells that produce high levels of TGF-beta. Sca-1 is also expressed by nearly all cells within fetal prostate epithelial chords, suggesting Sca-1 may be conserved on PSCs throughout development. Malignant epithelial cells from TRAMP mice, as well as normal prostate cells with lentiviral-mediated alteration of the PTEN/AKT signaling pathway, give rise to PIN lesions and prostate cancer in vivo. Alteration of PTEN/AKT signaling in Sca-1-enriched PSCs also results in PIN lesions, suggesting that PSCs can serve as one target for prostate carcinogenesis.
Collapse
Affiliation(s)
- D A Lawson
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, Howard Hughes Medical Institute, University of California, Los Angeles, 90095-1662, USA
| | | | | | | | | | | |
Collapse
|
234
|
Abstract
The hepatocyte growth factor/scatter factor (HGF/SF) and its receptor, the Met protein tyrosine kinase, form a classic ligand-receptor system for epithelial-mesenchymal communications in the normal and cancerous prostate. This review illustrates the expression and activities of HGF/SF and Met during prostate development, homeostasis, and carcinogenesis. The participation of HGF/SF in the morphogenetic program of rodent prostate development, the role of Met in normal human prostate epithelium, and underlying mechanisms of deregulated Met expression in localized and metastatic prostate cancer are discussed. On the basis of the commonly observed overexpression of Met in metastatic prostate cancer, HGF/SF-Met-targeted imaging and therapeutic agents can now be applied toward diagnosis and treatment.
Collapse
Affiliation(s)
- Beatrice S Knudsen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98125, USA
| | | |
Collapse
|
235
|
Abstract
The prostate contains two major epithelial cell types - luminal and basal cells - both of which develop from urogenital sinus epithelium. The cell linage relationship between these two epithelial types is not clear. Here we demonstrate that luminal cells can develop independently of basal cells, but that basal cells are essential for maintaining ductal integrity and the proper differentiation of luminal cells. Urogenital sinus (UGS) isolated from p63(+/+) and p63(-/-) embryos developed into prostate when grafted into adult male nude mice. Prostatic tissue that developed in p63(-/-) UGS grafts contained neuroendocrine and luminal cells, but basal cells were absent. Therefore, p63 is essential for differentiation of basal cells, but p63 and thus basal cells are not required for differentiation of prostatic neuroendocrine and luminal epithelial cells. p63(-/-) prostatic grafts also contained atypical mucinous cells, which appeared to differentiate from luminal cells via activation of Src. In the response to castration, regression of p63(-/-) prostate was inordinately severe with almost complete loss of ducts, resulting in the formation of residual cystic structures devoid of epithelium. Therefore, basal cells play critical roles in maintaining ductal integrity and survival of luminal cells. However, regressed p63(-/-) prostate did regenerate in response to androgen administration, indicating that basal cells were not essential for prostatic regeneration.
Collapse
Affiliation(s)
- Takeshi Kurita
- Department of Anatomy, University of California, San Francisco, CA 94143-0452, USA.
| | | | | | | |
Collapse
|
236
|
Alam TN, O'Hare MJ, Laczkó I, Freeman A, Al-Beidh F, Masters JR, Hudson DL. Differential expression of CD44 during human prostate epithelial cell differentiation. J Histochem Cytochem 2004; 52:1083-90. [PMID: 15258184 DOI: 10.1369/jhc.4a6256.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
CD44 is a polymorphic transmembrane glycoprotein that binds hyaluronan and growth factors. Multiple isoforms of the protein can be generated by alternative splicing but little is known about the expression and function of these isoforms in normal development and differentiation. We have investigated the expression of CD44 during normal prostate epithelial cell differentiation. A conditionally immortalized prostate epithelial cell line, Pre2.8, was used as a model system. These cells proliferate at 33C but at 39C stop dividing and undergo changes consistent with early stages of cell differentiation. During the differentiation of these cells, the expression of the CD44 isoform v3-v10 was upregulated. Two layers of epithelial cells can clearly be distinguished in the human prostate, a basal layer expressing keratins 5/14 and a luminal layer expressing keratins 8/18. In prostate tissue the v3-v10 isoform was found predominantly in basal cells but also in keratin 14-negative, keratin 19-positive cells intermediate between the two layers. CD44 v3-v10 was also expressed in other keratin 14-negative prostate tissues, the ejaculatory ducts and prostatic urethra. Therefore, CD44 v3-v10 may be important as a cell surface marker for differentiating cells in the prostate epithelium.
Collapse
Affiliation(s)
- Tahirah N Alam
- Prostate Cancer Research Centre, Institute of Urology, University College London, London, UK
| | | | | | | | | | | | | |
Collapse
|
237
|
Uzgare AR, Xu Y, Isaacs JT. In vitro culturing and characteristics of transit amplifying epithelial cells from human prostate tissue. J Cell Biochem 2004; 91:196-205. [PMID: 14689591 DOI: 10.1002/jcb.10764] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The prostatic epithelium is functionally organized in stem cell units. This unit consists of a slow turn over stem cell within the basal epithelial layer which can replenish itself and provide progeny which differentiate down either a neuroendocrine or exocrine pathway. The maturation along the exocrine pathway initially involves transit amplifying cells within the basal layer proliferating and subsequently the progeny maturing into intermediate cells. These intermediate cells migrate into the luminal layer where they terminally differentiate into non-proliferative secretory luminal cells which express prostate specific differentiation markers, like PSA. A growing body of experimental evidence has identified the proliferating transit amplifying/intermediate cells as the cells of origin for the common prostatic adenocarcinomas. Using a series of growth characteristics, and mRNA and protein markers, we have validated that primary cultures can be established in serum free defined media from surgically resected human prostates which are composed of essentially pure population of transit amplifying cells. At each serial passage, the subsequent cultures undergo enhanced maturation into intermediate cells and by the 7-10th passage these cells eventually lose their proliferative ability. This study validates that these cells are a useful and relevant system for the determination of molecular events involved in prostatic carcinogenesis.
Collapse
Affiliation(s)
- Aarti R Uzgare
- Division of Experimental Therapeutics, Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231, USA
| | | | | |
Collapse
|
238
|
Abstract
Benign prostatic hyperplasia and prostate cancer arise as a consequence of changes in the balance between cell division and differentiation. Little, however, is known about the control of this process. Stem cells are a small population of cells that divide occasionally to produce transit-amplifying cells that in turn produce the differentiated cell types of the tissue. It is believed that cancer cell proliferation is also driven by stem cells. We have shown that around one in 200 prostate epithelial cells have characteristics of stem cells and that these cells are contained within a population with a distinct keratin expression pattern. Work is now ongoing to identify markers for these cells that will allow us to study the role they play in prostatic disease.
Collapse
Affiliation(s)
- D L Hudson
- Bob Champion Prostate Stem Cell Laboratory, Male Urological Cancer Research Centre, Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| |
Collapse
|
239
|
Richardson GD, Robson CN, Lang SH, Neal DE, Maitland NJ, Collins AT. CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci 2004; 117:3539-45. [PMID: 15226377 DOI: 10.1242/jcs.01222] [Citation(s) in RCA: 551] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Stem cells are clonogenic cells with self-renewal and differentiation properties, which may represent a major target for genetic damage leading to prostate cancer and benign prostatic hyperplasia. Stem cells remain poorly characterised because of the absence of specific molecular markers that permit us to distinguish them from their progeny, the transit amplifying cells, which have a more restricted proliferative potential. Human CD133 antigen, also known as AC133, was recently identified as a haematopoietic stem cell marker. Here we show that a small population (approximately 1%) of human prostate basal cells express the cell surface marker CD133 and are restricted to the alpha(2)beta(1)(hi) population, previously shown to be a marker of stem cells in prostate epithelia. alpha(2)beta(1)(hi)/CD133(+) cells exhibit two important attributes of epithelial stem cells: they possess a high in vitro proliferative potential and can reconstitute prostatic-like acini in immunocompromised male nude mice.
Collapse
Affiliation(s)
- Gavin D Richardson
- Prostate Research Group, Surgical Oncology, Medical School, Framlington Place, University of Newcastle upon Tyne, Newcastle upon Tyne, NE2 4HH, UK
| | | | | | | | | | | |
Collapse
|
240
|
Islam TA, Patel A, Kaisary AV, Mondragon A, Waxman J, Kamalati T. In vitro deregulation of markers characteristic of human prostate epithelial cells. Tissue Cell 2004; 36:107-13. [PMID: 15041412 DOI: 10.1016/j.tice.2003.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2003] [Revised: 09/22/2003] [Accepted: 10/08/2003] [Indexed: 11/29/2022]
Abstract
We have screened primary cultures of human prostate for the expression of markers reported to be characteristic of specific cell lineages in vivo, in order to ascertain whether human prostate cells in vitro maintain and reflect their in vivo differentiated phenotypes and to evaluate the homogeneity of the populations of cells that can be derived from this tissue. Using single and dual stain immunofluorescent microscopy to analyse very early organoid and subsequently derived monolayer stage cultures, we have observed that expression of markers characteristic of human prostate epithelial cells in vivo is deregulated within 48h, indicating that dissociation of human prostate tissue and cultivation of prostate epithelial cells in culture can result in promiscuous expression of cell type specific markers of prostate epithelial cells. These observations have important implications for studies of cell lineage and differentiation of prostate cells in vitro.
Collapse
Affiliation(s)
- Tarin A Islam
- Cancer Cell Biology Section, Division of Medicine, Faculty of Medicine, Imperial College, Hammersmith Campus, 5th floor, Cyclotron Building, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | |
Collapse
|
241
|
Wang XD, Shou J, Wong P, French DM, Gao WQ. Notch1-expressing cells are indispensable for prostatic branching morphogenesis during development and re-growth following castration and androgen replacement. J Biol Chem 2004; 279:24733-44. [PMID: 15028713 DOI: 10.1074/jbc.m401602200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Notch expression is frequently associated with progenitor cells, and its function is crucial for development. Our recent work showing that Notch1 is selectively expressed in basal epithelial cells of the prostate and higher Notch1 expression during development suggests that Notch1-expressing cells may define progenitor cells in the prostate. To test this hypothesis, we have generated a transgenic mouse line in which the Notch1-expressing cells can be ablated in a controlled manner. Specific targeting was achieved by expressing the bacterial nitroreductase, an enzyme that catalyzes its substrate into a cytotoxin capable of inducing apoptosis, under the Notch1 promoter. Cell death in transgenic prostate was confirmed by histological analyses including terminal dUTP nick-end labeling and caspase 3 immunocytochemical staining. We evaluated the consequences of ablation of Notch1-expressing cells in two systems, organ culture of early postnatal prostates and re-growth of prostate in castrated mice triggered by hormone replacement. Our data show that elimination of Notch1-expressing cells inhibited the branching morphogenesis, growth, and differentiation of early postnatal prostate in culture and impaired prostate re-growth triggered by hormone replacement in castrated mice. Furthermore, we found that Notch1 expression following castration and hormone replacement was concomitant with known basal cell markers p63 and cytokeratin 14 and was high in the proliferative human prostate epithelial cells. Taken together, these data suggest that Notch1-expressing cells define the progenitor cells in the prostatic epithelial cell lineage, which are indispensable for prostatic development and re-growth.
Collapse
Affiliation(s)
- Xi-De Wang
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California 94080, USA
| | | | | | | | | |
Collapse
|
242
|
Xin L, Ide H, Kim Y, Dubey P, Witte ON. In vivo regeneration of murine prostate from dissociated cell populations of postnatal epithelia and urogenital sinus mesenchyme. Proc Natl Acad Sci U S A 2003; 100 Suppl 1:11896-903. [PMID: 12909713 PMCID: PMC304104 DOI: 10.1073/pnas.1734139100] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The existence of a postnatal prostate stem cell is supported by several types of evidence. Withdrawal of androgen leads to involution of the gland, but readdition can rapidly stimulate regeneration. Tissue fragments derived from mouse or rat prostatic epithelia from midgestation embryos or adult mice, when combined with tissue fragments from urogenital sinus mesenchyme and grafted under the kidney capsule, can regenerate prostatic structures. Indirect evidence supports that the stem cell population is contained within the basal layer. Purified prostatic stem cell preparations would be useful to define the physical and functional properties required for regeneration and to compare with cells that accumulate during abnormal growth states, like prostate cancer. We have developed a regeneration system using dissociated cell populations of postnatal prostate epithelia and embryonic urogenital sinus mesenchyme. Efficient in vivo regeneration of prostatic structures in the subcapsular space of the kidney was observed within 4-8 wk with as few as 103 epithelial cells from prostates derived from donors 10 d to 6 wk of age. The regenerated structures show a branching tubular epithelial morphology, with expression of a panel of markers consistent with prostate development. Donor epithelial populations can be readily infected with GFP expressing lentiviral vectors to provide integration markers and easy visualization. The cell preparations of urogenital sinus mesenchyme can be expanded in short-term in vitro culture while their inductive capabilities are retained. Further definition of the subpopulation of prostate epithelial cells containing the regeneration activity should be possible with such technologies.
Collapse
Affiliation(s)
- Li Xin
- Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | | | | | | | | |
Collapse
|
243
|
Bhatt RI, Brown MD, Hart CA, Gilmore P, Ramani VAC, George NJ, Clarke NW. Novel method for the isolation and characterisation of the putative prostatic stem cell. Cytometry A 2003; 54:89-99. [PMID: 12879455 DOI: 10.1002/cyto.a.10058] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Prostate stem cells, responsible for the development, maturation, and function of the prostate, have been implicated in the aetiology of both benign prostate hyperplasia (BPH) and prostate cancer (CaP). However, research has been hampered by the lack of a definitive stem cell marker. We have adapted the protocol for differential Hoechst 33342 uptake by hemopoietic stem cells to enable isolation of putative stem cells from the prostate. METHODS Prostate epithelial cells isolated from prostate tissue obtained from patients with BPH after transurethral resection of the prostate were stained with Hoechst 33342. The Hoechst 33342 Red/Blue flow cytometry profile was then determined. Hoechst 33342 and Pyronin Y staining was used to determined the cell cycle status. RESULTS A verapamil-sensitive side population (SP) can be isolated from primary prostate tissue accounting for 1.38% +/- 0.07% of prostate epithelial cells. Cell cycle analysis of this SP population revealed that the majority of SP cells are in either G0 (12.38 +/- 0.31%) or G1 (63.19 +/- 2.13%). CONCLUSIONS The Hoechst 33342 dye efflux protocol can be adapted for the isolation of a SP from primary prostate tissue.
Collapse
Affiliation(s)
- Rupesh I Bhatt
- Genito-Urinary Cancer Research Group, Cancer Research UK Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester, UK
| | | | | | | | | | | | | |
Collapse
|
244
|
Keay S, Seillier-Moiseiwitsch F, Zhang CO, Chai TC, Zhang J. Changes in human bladder epithelial cell gene expression associated with interstitial cystitis or antiproliferative factor treatment. Physiol Genomics 2003; 14:107-15. [PMID: 12847144 DOI: 10.1152/physiolgenomics.00055.2003] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Explanted bladder epithelial cells from patients with interstitial cystitis (IC) have been shown to differ from explanted control cells in several ways, including production of an antiproliferative factor (APF), altered production of certain epithelial growth factors, and rate of proliferation. To better understand the role of the APF in abnormal bladder epithelial cell proliferation in IC, we studied gene expression patterns in normal bladder epithelial cells treated with APF vs. mock APF and compared them to expression patterns in IC vs. normal cells using microarray analysis. Oligo-dT-primed total cellular RNA was labeled with [(33)P]dCTP and hybridized to GeneFilter GF211 microarray membranes (Research Genetics) containing cDNA for 3,964 human genes. Thirteen genes that function in epithelial cell proliferation or differentiation were consistently differentially expressed in both IC (compared with control) and APF-treated (compared with mock APF-treated) normal bladder epithelial cells. The general pattern of gene expression in IC and APF-treated cells suggested a less proliferative phenotype, with increased expression of E-cadherin, phosphoribosylpyrophosphate synthetase-associated protein 39, and SWI/SNF complex 170-kDa subunit, and decreased expression of vimentin, alpha2-integrin, alpha1-catenin, cyclin D1, and jun N-terminal kinase 1; these findings were confirmed for the structural gene products (E-cadherin, vimentin, alpha2-integrin, and alpha-catenin) by immunohistochemistry. These results are compatible with the previously noted decreased proliferation rate of IC and APF-treated normal cells, and indicate that the mechanism whereby APF inhibits cell proliferation may involve both downregulation of genes that stimulate cell proliferation along with upregulation of genes that inhibit cell growth.
Collapse
Affiliation(s)
- Susan Keay
- Division of Infectious Diseases, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | |
Collapse
|
245
|
Grieco V, Patton V, Romussi S, Finazzi M. Cytokeratin and vimentin expression in normal and neoplastic canine prostate. J Comp Pathol 2003; 129:78-84. [PMID: 12859911 DOI: 10.1016/s0021-9975(03)00006-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Intermediate filament expression in the canine prostate, unlike that in human prostate, is represented in the literature by only a few reports. In this study, the expression of cytokeratin (CK) and vimentin was examined in three normal canine prostates and 11 canine prostatic carcinomas. Monoclonal antibodies directed against vimentin, CK AE1/AE3, CK 18-8 (for luminal epithelial cells), CK 5, CK clone 8.12 and CK 14 (for basal cells) were employed. As in man, normal canine prostatic luminal cells were positive for CK 8-18. Basal cells were positive for CK 5 and CK clone 8.12 but, in contrast to findings in man, were negative for CK 14. Luminal cells were vimentin-negative, whereas in man they have been reported as vimentin-positive. The majority of carcinomas showed an undifferentiated histological pattern and all were positive for CK AE1/AE3 and for vimentin. Ten tumours were positive for CK 8-12, but six of them showed many cells co-expressing CK 14. Moreover, in two of these six cases a large number of neoplastic cells also reacted with CK clone 8.12 antibody, and in one of them co-expression of CK 5 was detectable. This co-expression, of luminal and basal cytokeratins, suggests a possible origin of the tumours from prostatic epithelial stem cells. Vimentin expression is an inconstant finding in human prostatic carcinomas; its almost uniform occurrence in canine carcinomas suggests a lesser degree of differentiation than in the human neoplasm.
Collapse
Affiliation(s)
- V Grieco
- Dipartimento di Patologia, Igiene e Sanità Pubblica Veterinaria, Sezione di Anatomia Patologica e Patologia Aviare, Facoltà di Medicina Veterinaria, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | | | | | | |
Collapse
|
246
|
van Leenders GJLH, Schalken JA. Epithelial cell differentiation in the human prostate epithelium: implications for the pathogenesis and therapy of prostate cancer. Crit Rev Oncol Hematol 2003; 46 Suppl:S3-10. [PMID: 12850522 DOI: 10.1016/s1040-8428(03)00059-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Within the human prostate epithelium four cell populations are discriminated by their expression of keratins (K). While basal cells co-localize K5 and K14 combined with low levels of K18 (K5(++)/14(++)/18(+)), luminal cells highly express K18 (K18(++)). In addition, two intermediate subpopulations are characterized either by basal K5(++)/18(+)- or luminal K5(+)/18(++)- expression. The entire prostate epithelium is putatively derived from a basal stem cell population. They give rise to intermediate cells that transiently proliferate and mature towards differentiated luminal epithelium. Within prostate carcinoma luminal exocrine, neuro-endocrine and intermediate cells are distinguished. Intermediate cells have been postulated as progenitors for prostate carcinogenesis and targets for androgen-independent tumor progression. Androgen-independency is associated with an enrichment of intermediate cells and over-expression of peptide growth factor receptors. Targeting intermediate cells by inhibition of their peptide growth factor receptors, therefore, offers novel treatment modalities for prostate cancer.
Collapse
Affiliation(s)
- Geert J L H van Leenders
- Department of Pathology, University Medical Center 'St. Radboud', P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | | |
Collapse
|
247
|
Marker PC, Donjacour AA, Dahiya R, Cunha GR. Hormonal, cellular, and molecular control of prostatic development. Dev Biol 2003; 253:165-74. [PMID: 12645922 DOI: 10.1016/s0012-1606(02)00031-3] [Citation(s) in RCA: 345] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The prostate is a male accessory sex gland found only in mammals that functions to produce a major fraction of seminal fluid. Interest in understanding the biology of the prostate is driven both by the fascinating nature of the developmental processes that give rise to the prostate and by the high incidence in humans of prostatic diseases, including prostatic adenocarcinoma and benign prostatic hyperplasia. This review summarizes the current state of knowledge of the cellular and molecular processes that control prostatic development. Insight into the mechanisms that control prostatic development has come from experimental embryological work as well as from the study of mice and humans harboring mutations that alter prostatic development. These studies have demonstrated a requirement for androgens throughout prostatic development and have revealed a series of reciprocal paracrine signals between the developing prostatic epithelium and prostatic mesenchyme. Finally, these studies have identified several specific gene products that are required for prostatic development. While research in recent years has greatly enhanced our understanding of the molecular control of prostatic development, known genes cannot yet explain in molecular terms the complex biological interactions that descriptive and experimental embryological studies have elucidated in the control of prostatic development.
Collapse
Affiliation(s)
- Paul C Marker
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
248
|
Rumpold H, Heinrich E, Untergasser G, Hermann M, Pfister G, Plas E, Berger P. Neuroendocrine differentiation of human prostatic primary epithelial cells in vitro. Prostate 2002; 53:101-8. [PMID: 12242724 DOI: 10.1002/pros.10129] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Dispersed prostatic neuroendocrine cells are involved in growth regulation of the prostate and are considered to play a role in the pathogenesis of prostate carcinoma and benign prostatic hyperplasia (BPH). They are meant either to be derived from the neural crest during embryogenesis or by direct differentiation of the cells from locally present precursor cells. METHODS An in vitro model was developed for human prostatic epithelial and neuroendocrine cell differentiation. Minced explants from radical prostatectomies were seeded on collagen I-coated plates. RESULTS The majority of outgrowing cells were basal cells, positive for cytokeratin markers K 5/14 and CD 44, as determined by confocal laser scanning microscopy. A small fraction of interdispersed single cells expressing c-kit, which is found on pluripotent precursors, was identified by immunofluorescence. From these basal cells, in vitro differentiation of cells with neuroendocrine morphology could be achieved within 3 days. These were at rest, i.e., non-bromodeoxyuridine incorporating cells and characteristically coexpressed K 5/14, K 18, and the neuroendocrine marker chromogranin A. Luminal cells staining for K 8 or 18 were not observed. CONCLUSION Neuroendocrine differentiation of adult prostatic cells was achieved in vitro, favoring the hypothesis that neuroendocrine cells are derived from peripheral precursor cells. The acceleration of this differentiation pathway may be the reason for the increased presence of neuroendocrine cells in areas of epithelial hyperplasia in BPH.
Collapse
Affiliation(s)
- Holger Rumpold
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
249
|
Rumpold H, Untergasser G, Madersbacher S, Berger P. The development of benign prostatic hyperplasia by trans-differentiation of prostatic stromal cells. Exp Gerontol 2002; 37:1001-4. [PMID: 12213550 DOI: 10.1016/s0531-5565(02)00062-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- H Rumpold
- Austrian Academy of Sciences, Institute for Biomedical Aging Research, Peter-Mayr-Street 4b, Innsbruck A6020, Austria
| | | | | | | |
Collapse
|