Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 38(29): 5766-5777, 2019 07.
Article in English | MEDLINE | ID: mdl-31239516

ABSTRACT

We previously showed that KLF4, a gene highly expressed in murine prostate stem cells, blocks the progression of indolent intraepithelial prostatic lesions into aggressive and rapidly growing tumors. Here, we show that the anti-tumorigenic effect of KLF4 extends to PC3 human prostate cancer cells growing in the bone. We compared KLF4 null cells with cells transduced with a DOX-inducible KLF4 expression system, and find KLF4 function inhibits PC3 growth in monolayer and soft agar cultures. Furthermore, KLF4 null cells proliferate rapidly, forming large, invasive, and osteolytic tumors when injected into mouse femurs, whereas KLF4 re-expression immediately after their intra-femoral inoculation blocks tumor development and preserves a normal bone architecture. KLF4 re-expression in established KLF4 null bone tumors inhibits their osteolytic effects, preventing bone fractures and inducing an osteogenic response with new bone formation. In addition to these profound biological changes, KLF4 also induces a transcriptional shift from an osteolytic program in KLF4 null cells to an osteogenic program. Importantly, bioinformatic analysis shows that genes regulated by KLF4 overlap significantly with those expressed in metastatic prostate cancer patients and in three individual cohorts with bone metastases, strengthening the clinical relevance of the findings in our xenograft model.


Subject(s)
Bone Neoplasms/secondary , Kruppel-Like Transcription Factors/physiology , Osteolysis/physiopathology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cohort Studies , Heterografts , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
2.
Cell Rep ; 25(11): 3006-3020.e7, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540935

ABSTRACT

There is a considerable need to identify those individuals with prostate cancer who have indolent disease. We propose that genes that control adult stem cell homeostasis in organs with slow turnover, such as the prostate, control cancer fate. One such gene, KLF4, overexpressed in murine prostate stem cells, regulates their homeostasis, blocks malignant transformation, and controls the self-renewal of tumor-initiating cells. KLF4 loss induces the molecular features of aggressive cancer and converts PIN lesions to invasive sarcomatoid carcinomas; its re-expression in vivo reverses this process. Bioinformatic analysis links these changes to human cancer. KLF4 and its downstream targets make up a gene signature that identifies indolent tumors and predicts recurrence-free survival. This approach may improve prognosis and identify therapeutic targets for advanced cancer.


Subject(s)
Disease Progression , Gene Expression Regulation, Neoplastic , Homeostasis , Kruppel-Like Transcription Factors/genetics , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cell Self Renewal/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Phenotype , Prognosis
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.
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
5.
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
6.
J Cell Biol ; 176(6): 787-93, 2007 Mar 12.
Article in English | MEDLINE | ID: mdl-17353357

ABSTRACT

The multifunctional cytokine transforming growth factor (TGF) beta1 is secreted in a latent complex with its processed propeptide (latency-associated peptide [LAP]). TGFbeta1 must be functionally released from this complex before it can engage TGFbeta receptors. One mechanism of latent TGFbeta1 activation involves interaction of the integrins alpha v beta6 and alpha v beta8 with an RGD sequence in LAP; other putative latent TGFbeta1 activators include thrombospondin-1, oxidants, and various proteases. To assess the contribution of RGD-binding integrins to TGFbeta1 activation in vivo, we created a mutation in Tgfb1 encoding a nonfunctional variant of the RGD sequence (RGE). Mice with this mutation (Tgfb1(RGE/RGE)) display the major features of Tgfb1(-/-) mice (vasculogenesis defects, multiorgan inflammation, and lack of Langerhans cells) despite production of normal levels of latent TGFbeta1. These findings indicate that RGD-binding integrins are requisite latent TGFbeta1 activators during development and in the immune system.


Subject(s)
Integrins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Binding Sites , Mice , Mice, Knockout , Phenotype , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/genetics , Vascular Diseases/genetics , Vascular Diseases/pathology , Yolk Sac/blood supply , Yolk Sac/pathology
7.
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
8.
J Leukoc Biol ; 79(1): 140-6, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16244108

ABSTRACT

Systemic injection of small amounts of transforming growth factor-beta (TGF-beta), a cytokine produced by lymphoid and other cells, has a profound effect in protecting mice from the inflammatory demyelinating lesions of experimental allergic encephalomyelitis (EAE; an animal model for multiple sclerosis). However, TGF-beta has side-effects, which might be avoided if the cells producing TGF-beta can be delivered to the affected site in the nervous system to insure its local release in small amounts. Myelin basic protein (MBP)-specific, cloned CD4+ T cells were engineered by retroviral transduction to produce latent TGF-beta. Studies about the spontaneous form of EAE in T cell receptor (TCR)-transgenic recombination-activating gene (RAG)-1(-/-) mice showed that essentially all of the MBP-specific, TCR-transgenic RAG-1(-/-) (BALB/cxB10.PL)F1 mice develop spontaneous EAE by the age of 11 weeks. By 12 weeks, 25-50% of the mice have died from disease. A single injection of TGF-beta1-transduced T helper cell type 1 (Th1) cells significantly protected the mice from EAE, and untransduced Th1 cells did not protect. MBP-specific BALB/c Th2 clones, transduced with TGF-beta1-internal ribosome entry site-green fluorescent protein (GFP) significantly reduced EAE induction by untransduced Th1 cells in RAG-1(-/-) B10.PL mice. Furthermore, the GFP+ TGF-beta1-producing Th2 cells were detectable in the spinal cords of the injected mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin Basic Protein/immunology , Th1 Cells/immunology , Transforming Growth Factor beta/immunology , Adoptive Transfer , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/therapy , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Myelin Basic Protein/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Th1 Cells/transplantation , Th2 Cells/immunology , Transduction, Genetic , Transforming Growth Factor beta/administration & dosage , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
9.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...