Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Res ; 11(6): 604-15, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23525267

ABSTRACT

Although Hedgehog signaling plays a major role in GLI1 transcription, there is now evidence suggesting that other pathways/genes, such as c-MYC, may also regulate GLI1 expression. We initiated studies in Burkitt lymphoma cells, which constitutively express c-MYC due to a chromosomal translocation, to determine whether Hedgehog or c-MYC regulates GLI1 expression. We show that all Burkitt lymphoma cell lines tested express GLI1, PTCH1, and SMO and that five of six Burkitt lymphomas express GLI1. Exposure to Sonic or Indian Hedgehog or cyclopamine (SMO inhibitor) does not modulate GLI1 expression, cell proliferation, or apoptosis in most Burkitt lymphoma cell lines. Sequence analysis of PTCH1, SMO, and SuFu failed to show mutations that might explain the lack of Hedgehog responsiveness, and we did not detect primary cilia, which may contribute to it. We show that c-MYC interacts with the 5'-regulatory region of GLI1, using chromatin immunoprecipitation (ChIP) assay, and E-box-dependent transcriptional activation of GLI1 by c-MYC in NIH3T3 and HeLa cells. The c-MYC small-molecule inhibitor 10058-F4 downregulates GLI1 mRNA and protein and reduces the viability of Burkitt lymphoma cells. Inhibition of GLI1 by GANT61 increases apoptosis and reduces viability of some Burkitt lymphoma cells. Collectively, our data provide evidence that c-MYC directly regulates GLI1 and support an antiapoptotic role for GLI1 in Burkitt lymphoma. Burkitt lymphoma cells do not seem to be Hedgehog responsive. These findings suggest a mechanism for resistance to SMO inhibitors and have implications for using SMO inhibitors to treat human cancers.


Subject(s)
Burkitt Lymphoma/metabolism , Hedgehog Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , 5' Untranslated Regions/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Cell Line, Tumor , Cilia/drug effects , Cilia/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/genetics , Hedgehog Proteins/pharmacology , Humans , Mice , NIH 3T3 Cells , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1
2.
Mol Cancer ; 11: 30, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22559324

ABSTRACT

BACKGROUND: Induction of osteoblast differentiation by paracrine Sonic hedgehog (Shh) signaling may be a mechanism through which Shh-expressing prostate cancer cells initiate changes in the bone microenvironment and promote metastases. A hallmark of osteoblast differentiation is the formation of matrix whose predominant protein is type 1 collagen. We investigated the formation of a collagen matrix by osteoblasts cultured with prostate cancer cells, and its effects on interactions between prostate cancer cells and osteoblasts. RESULTS: In the presence of exogenous ascorbic acid (AA), a co-factor in collagen synthesis, mouse MC3T3 pre-osteoblasts in mixed cultures with human LNCaP prostate cancer cells or LNCaP cells modified to overexpress Shh (LNShh cells) formed collagen matrix with distinct fibril ultrastructural characteristics. AA increased the activity of alkaline phosphatase and the expression of the alkaline phosphatase gene Akp2, markers of osteoblast differentiation, in MC3T3 pre-osteoblasts cultured with LNCaP or LNShh cells. However, the AA-stimulated increase in Akp2 expression in MC3T3 pre-osteoblasts cultured with LNShh cells far exceeded the levels observed in MC3T3 cells cultured with either LNCaP cells with AA or LNShh cells without AA. Therefore, AA and Shh exert a synergistic effect on osteoblast differentiation. We determined whether the effect of AA on LNShh cell-induced osteoblast differentiation was mediated by Shh signaling. AA increased the expression of Gli1 and Ptc1, target genes of the Shh pathway, in MC3T3 pre-osteoblasts cultured with LNShh cells to at least twice their levels without AA. The ability of AA to upregulate Shh signaling and enhance alkaline phosphatase activity was blocked in MC3T3 cells that expressed a dominant negative form of the transcription factor GLI1. The AA-stimulated increase in Shh signaling and Shh-induced osteoblast differentiation was also inhibited by the specific collagen synthesis inhibitor 3,4-dehydro-L-proline. CONCLUSIONS: Matrix collagen, formed by osteoblasts in the presence of AA, potentiates Shh signaling between Shh-expressing prostate cancer cells and osteoblasts. Collagen and Shh signaling exert a synergistic effect on osteoblast differentiation, a defining event in prostate carcinoma bone metastasis. Investigations into paracrine interactions among prostate cancer cells, osteoblasts, and osteoblast-synthesized matrix proteins advance our understanding of mechanisms contributing to prostate cancer bone metastasis.


Subject(s)
Cell Differentiation/physiology , Fibrillar Collagens/metabolism , Hedgehog Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Paracrine Communication/physiology , Prostatic Neoplasms/metabolism , Alkaline Phosphatase/metabolism , Animals , Ascorbic Acid/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Coculture Techniques , Fibrillar Collagens/chemistry , Histocytochemistry , Humans , Male , Mice , Paracrine Communication/drug effects , Particle Size , Prostatic Neoplasms/pathology , Up-Regulation
3.
Mol Cancer ; 8: 12, 2009 Mar 02.
Article in English | MEDLINE | ID: mdl-19254376

ABSTRACT

BACKGROUND: Sonic hedgehog (Shh) and components of its signalling pathway have been identified in human prostate carcinoma and increased levels of their expression appear to correlate with disease progression and metastasis. The mechanism through which Shh signalling could promote metastasis in bone, the most common site for prostate carcinoma metastasis, has not yet been investigated. The present study determined the effect of Shh signalling between prostate cancer cells and pre-osteoblasts on osteoblast differentiation, a requisite process for new bone formation that characterizes prostate carcinoma metastasis. RESULTS: LNCaP human prostate cancer cells modified to overexpress Shh (designated LNShh cells) and MC3T3 mouse pre-osteoblasts were maintained as mixed populations within the same culture chamber. In this non-conventional mixed culture system, LNShh cells upregulated the expression of Shh target genes Gli1 and Patched 1 (Ptc1) in MC3T3 cells and this was inhibited by cyclopamine, a specific chemical inhibitor of hedgehog signalling. Concomitantly, MC3T3 cells exhibited time-dependent decreased cell proliferation, upregulated alkaline phosphatase Akp2 gene expression, and increased alkaline phosphatase activity indicative of early phase osteoblast differentiation. LNShh cell-induced differentiation was inhibited in MC3T3 cells stably transfected with a dominant negative form of Gli1, a transcription factor that mediates Shh signalling. Interestingly, LNShh cells did not significantly increase the endogenous expression of the osteoblast differentiation transcription factor Runx2 and its target genes osteocalcin and osteopontin. Consistent with these results, exogenous Shh peptide did not upregulate Runx2 expression in MC3T3 cells. However, Runx2 levels were increased in MC3T3 cells by ascorbic acid, a known stimulator of osteoblast differentiation. CONCLUSION: Altogether, these data demonstrate that Shh-expressing prostate cancer cells can directly and specifically induce differentiation in pre-osteoblasts via a Gli1-dependent mechanism that does not require transcriptional upregulation of Runx2. Paracrine activation of the Shh pathway in osteoblast progenitors and subsequent induction of osteoblast differentiation could be a mechanism through which high levels of Shh expression in prostate carcinoma contribute to bone metastasis. Targeting of paracrine Shh signalling may provide an effective therapeutic strategy against prostate carcinoma metastasis in bone.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Osteoblasts/cytology , Paracrine Communication , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/genetics , Genes, Dominant , Humans , Male , Mice , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/metabolism , Transcription Factors/genetics , Transcription, Genetic , Up-Regulation/genetics , Zinc Finger Protein GLI1
4.
Endocrinology ; 145(8): 3961-70, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15132968

ABSTRACT

During fetal prostate development, Sonic hedgehog (Shh) expression by the urogenital sinus epithelium activates Gli-1 expression in the adjacent mesenchyme and promotes outgrowth of the nascent ducts. Shh signaling is down-regulated at the conclusion of prostate ductal development. However, a survey of adult human prostate tissues reveals substantial levels of Shh signaling in normal, hyperplasic, and malignant prostate tissue. In cancer specimens, the Shh expression is localized to the tumor epithelium, whereas Gli-1 expression is localized to the tumor stroma. Tight correlation between the levels of Shh and Gli-1 expression suggests active signaling between the tissue layers. To determine whether Shh-Gli-1 signaling could be functionally important for tumor growth and progression, we performed experiments with the LNCaP xenograft tumor model and demonstrated that: 1). Shh expressed by LNCaP tumor cells activates Gli-1 expression in the tumor stroma, 2). genetically engineered Shh overexpression in LNCaP cells leads to increased tumor stromal Gli-1 expression, and 3). Shh overexpression dramatically accelerates tumor growth. These data suggest that hedgehog signaling from prostate cancer cells to the stroma can elicit the expression of paracrine signals, which promote tumor growth.


Subject(s)
Prostatic Neoplasms/pathology , Signal Transduction , Trans-Activators/physiology , Adult , Animals , Cell Line, Tumor , Hedgehog Proteins , Humans , Intracellular Signaling Peptides and Proteins , Kruppel-Like Transcription Factors , Male , Membrane Proteins , Mice , Neoplasm Transplantation , Patched Receptors , Prostatic Neoplasms/metabolism , Proteins/physiology , Receptors, Cell Surface , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/physiology , Transplantation, Heterologous , Zinc Finger Protein GLI1
5.
Biol Reprod ; 69(1): 8-14, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12672657

ABSTRACT

Hedgehog (Hh) proteins are expressed during vertebrate development in some tissues with inductive properties and at epithelial-mesenchymal boundaries in several developing organs, including the lung, gut, hair follicle, and tooth. The Hh signaling pathway is highly conserved, and important clues to understanding the mechanism of Hh signal transduction in vertebrates have come from studies in Drosophila. In recent years, Hh signaling has been recognized during embryonic development and in some cases during postnatal life in several mammalian tissues whose functions are essential for reproduction, including the gonads, uterus, and hormonally responsive accessory sex glands such as the prostate and mammary gland. The role of the pathway in these tissues is highly reminiscent of its role at epithelial-mesenchymal-stromal boundaries in other organ systems, which has provided a framework within which to explore Hh signaling in tissues that function in reproduction. Some features unique to these tissues are emerging, including a role in proliferation and differentiation of male germline cells in mammals and apparent influences of sex steroids on Hh signaling. However, many questions remain about the function of Hh signaling in the gonads, uterus, prostate, and mammary gland, including factors regulating the signal transduction pathway, identification of downstream target genes, and roles for Hh signaling in diseases involving these tissues.


Subject(s)
Reproduction/physiology , Animals , Drosophila , Drosophila Proteins/physiology , Female , Genitalia, Female/physiology , Genitalia, Male/physiology , Hedgehog Proteins , Male , Membrane Proteins/physiology , Oncogene Proteins/physiology , Patched Receptors , Pregnancy , Receptors, Cell Surface , Signal Transduction , Trans-Activators/physiology , Transcription Factors/physiology , Vertebrates , Zinc Finger Protein GLI1
SELECTION OF CITATIONS
SEARCH DETAIL
...