Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Pigment Cell Melanoma Res ; 24(6): 1133-47, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21978367

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) have been shown to regulate signaling in many systems and are of increasing interest in cancer. While these are not the only sugars to drive melanoma metastasis, HSPGs play important roles in driving metastatic signaling cascades in melanoma. The ability of these proteins to modulate ligand-receptor interactions in melanoma has been quite understudied. Recent data from several groups indicate the importance of these ligands in modulating key signaling pathways including Wnt and fibroblast growth factor (FGF) signaling. In this review, we summarize the current knowledge regarding the structure and function of these proteoglycans and their role in melanoma. Understanding how HSPGs modulate signaling in melanoma could lead to new therapeutic approaches via the dampening or heightening of key signaling pathways.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Melanoma/pathology , Skin Neoplasms/pathology , Animals , Heparan Sulfate Proteoglycans/antagonists & inhibitors , Humans , Melanoma/metabolism , Melanoma/therapy , Molecular Targeted Therapy , Neoplasm Metastasis , Skin Neoplasms/therapy
2.
Pigment Cell Melanoma Res ; 24(1): e1-15, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21232023

ABSTRACT

The 2010 7th International Melanoma Congress sponsored by the Society for Melanoma Research and held in Sydney, Australia, was held together with the International Melanoma and Skin Cancer Centers group and the International Melanoma Pathology Study Group. As a consequence, there were over 900 registrants that included a wide range of clinicians (surgeons, medical oncologists, dermatologists) specialising in the management of melanoma as well as scientists and students carrying out laboratory-based research in melanoma. There was a general consensus that this grouping of clinicians, pathologists and scientists was mutually advantageous and plans are afoot to continue this grouping in future meetings. The meeting was dominated by the advances being made in treatment of melanoma with selective BRAF inhibitors but interest in epithelial mesenchymal transition and phenotypic changes in melanoma was apparent in many of the talks. The authors have attempted to capture many of the new developments in melanoma research but apologize to those speakers and poster presenters who had equally important findings not captured in these summaries.


Subject(s)
Congresses as Topic , Melanoma/pathology , Animals , Clinical Trials as Topic , Disease Models, Animal , Drug Screening Assays, Antitumor , Humans , Immunotherapy , Melanoma/drug therapy , Melanoma/genetics , Melanoma/therapy , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , New South Wales , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Zebrafish/genetics
3.
Oncogene ; 29(1): 34-44, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-19802008

ABSTRACT

Tyrosine kinase receptors represent targets of great interest for cancer therapy. Here we show, for the first time, the importance of the orphan tyrosine kinase receptor, ROR2, in melanoma progression. Using melanoma tissue microarrays, we show that ROR2 is expressed predominantly in metastatic melanoma. As ROR2 has been shown to specifically interact with the non-canonical Wnt ligand, Wnt5A, this corroborates our earlier data implicating Wnt5A as a mediator of melanoma metastasis. We show here that increases in Wnt5A cause increases in ROR2 expression, as well as the PKC-dependent, clathrin-mediated internalization of ROR2. WNT5A knockdown by siRNA decreases ROR2 expression, but silencing of ROR2 has no effect on WNT5A levels. ROR2 knockdown does, however, result in a decrease in signaling downstream of Wnt5A. Using in vitro and in vivo metastasis assays, we show that ROR2 is necessary for the Wnt5A-mediated metastasis of melanoma cells. These data imply that ROR2 may represent a novel target for melanoma therapy.


Subject(s)
Melanoma/pathology , Proto-Oncogene Proteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Signal Transduction , Wnt Proteins/metabolism , Aged , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement , Endocytosis/drug effects , Humans , Immunohistochemistry , Melanoma/genetics , Melanoma/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis , Proto-Oncogene Proteins/genetics , RNA Interference , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Wnt Proteins/genetics , Wnt-5a Protein
4.
Oncogene ; 26(26): 3846-56, 2007 May 31.
Article in English | MEDLINE | ID: mdl-17160014

ABSTRACT

Serial analysis of gene expression followed by pathway analysis implicated the tight junction protein claudin-1 (CLDN1) in melanoma progression. Tight junction proteins regulate the paracellular transport of molecules, but staining of a tissue microarray revealed that claudin-1 was overexpressed in melanoma, and aberrantly expressed in the cytoplasm of malignant cells, suggesting a role other than transport. Indeed, melanoma cells in culture demonstrate no tight junction function. It has been shown that protein kinase C (PKC) can affect expression of claudin-1 in rat choroid plexus cells, and we observed a correlation between levels of activated PKC and claudin expression in our melanoma cells. To determine if PKC could affect the expression of CLDN1 in human melanoma, cells lacking endogenous claudin-1 were treated with 200 nM phorbol myristic acid (PMA). PKC activation by PMA caused an increase in CLDN1 transcription in 30 min, and an increase in claudin-1 protein by 12 h. Inhibition of PKC signaling in cells with high claudin-1 expression resulted in decreased claudin-1 expression. CLDN1 appears to contribute to melanoma cell invasion, as transient transfection of melanoma cells with CLDN1 increased metalloproteinase 2 (MMP-2) secretion and activation, and subsequently, motility of melanoma cells as demonstrated by wound-healing assays. Conversely, knockdown of CLDN1 by siRNA resulted in the inhibition of motility, as well as decreases in MMP-2 secretion and activation. These data implicate claudin-1 in melanoma progression.


Subject(s)
Cell Movement/physiology , Melanoma/pathology , Membrane Proteins/metabolism , Neoplasm Invasiveness/physiopathology , Protein Kinase C/metabolism , Blotting, Western , Cell Line, Tumor , Claudin-1 , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Matrix Metalloproteinase 2/metabolism , Melanoma/metabolism , Microscopy, Confocal , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection
5.
Oncogene ; 26(20): 2860-72, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17072341

ABSTRACT

The molecular pathways activated in response to acute cisplatin exposure, as well as the mechanisms involved in the long-term development of cisplatin-resistant cancer cells remain unclear. Using whole genome oligonucleotide microarrays, we have examined the kinetics of gene expression changes in a cisplatin-sensitive cell line, A2780, and its cisplatin-resistant derivative, ACRP. Both sensitive and resistant cell lines exhibited a very similar response of p53-inducible genes as early as 16 h after treatment. This p53 response was further increased at the 24-h time point. These experiments identify p53 as the main pathway producing a large-scale transcriptional response after cisplatin treatment in these cells containing wild-type p53. Consistent with a role for the p53 response in cisplatin sensitivity, knockdown of the p53 protein with small interfering RNA led to a twofold decrease in cell survival in the resistant cells. In addition, our analysis also allowed the identification of several genes that are differentially expressed between sensitive and resistant cells. These genes include GJA1 (encoding connexin 43 (Cx43)) and TWIST1, which are highly upregulated in cisplatin-resistant cells. The importance of Cx43 in drug resistance was demonstrated through functional analyses, although paradoxically, inhibition of Cx43 function in high expressing cells led to an increase in drug resistance. The pathways important in cisplatin response, as well as the genes found differentially expressed between cisplatin-resistant and -sensitive cells, may represent targets for therapy aimed at reversing drug resistance.


Subject(s)
Cisplatin/pharmacology , Cystadenocarcinoma, Serous/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Ovarian Neoplasms/genetics , Cluster Analysis , Connexin 43/genetics , Cystadenocarcinoma, Serous/pathology , Female , Gene Expression Profiling , Humans , Ovarian Neoplasms/pathology , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors
6.
Oncogene ; 25(40): 5561-9, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16619041

ABSTRACT

Transforming growth factor-beta type 1 (TGF-beta) has been implicated as both a tumor suppressor and a tumor promoter in many solid epithelial cancers. We have previously demonstrated that the cyclin dependent kinase (CDK) inhibitor p21 acts as a molecular switch in determining a growth inhibitory versus growth proliferative response to TGF-beta in the spontaneously immortalized human mammary epithelial cell line MCF-10A. We now demonstrate that this proliferative effect of TGF-beta is mediated through the proinflammatory cytokine, interleukin-1alpha (IL-1alpha). Using gene expression array analysis, we identified IL-1alpha as a cytokine specifically upregulated only in cells lacking p21 and only upon TGF-beta stimulation. Cell proliferation assays verified that recombinant IL-1alpha was capable of inducing a growth proliferative response in p21 null MCF-10A cells, while neutralizing antibodies against IL-1alpha prevented the growth proliferative effects of TGF-beta. Mechanistically, both the CDK and proliferating cell nuclear antigen (PCNA) inhibitory functions of p21 were responsible for preventing TGF-beta induced cell proliferation, but only PCNA inhibition by p21 regulated IL-1alpha gene expression. These studies demonstrate a novel role for IL-1alpha in mediating a proliferative response to TGF-beta signaling, and suggest that therapies directed against IL-1alpha could abate the growth proliferative effects of TGF-beta without compromising its tumor suppressive function.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Interleukin-1/metabolism , Second Messenger Systems , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dactinomycin/pharmacology , Humans , Interleukin-1/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Oligonucleotide Array Sequence Analysis , Proliferating Cell Nuclear Antigen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Up-Regulation
7.
Clin Cancer Res ; 7(8): 2237-45, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489797

ABSTRACT

During the progression of prostate cancer, molecular changes occur resulting in the autocrine production of a series of neurotrophins by the malignant cells. This is coupled with expression of high-affinity cognate receptors for these ligands, termed trk receptors, by these cancer cells. The binding of the neurotrophins to their trk receptors activates the receptor's latent tyrosine kinase activity inducing a series of signal transduction pathways within these prostate cancer cells. These molecular changes result in the acquisition by prostate cancer cells of a restricted requirement for these trk signaling pathways for optimal survival. CEP-701 is an indolocarbazole compound specifically designed as a potent inhibitor (IC(50), 4 nM) of the tyrosine kinase activity of the trk receptors required for initiation of these survival pathways. In the present studies, the consequences of CEP-701 inhibition of these trk signaling survival pathways were tested in vivo using both rat (R3327 AT 6.3 and H) and human (TSU-pr1 and CWR-22Rv1) prostatic cancer models. These in vivo studies demonstrated that treatment with CEP-701 inhibits the growth of both rodent and human prostate cancers, without being toxic to the normal tissue including the host prostate. Because of this selective effect, CEP-701 inhibits metastasis and growth of both primary and metastatic sites of prostate cancer. Based upon this profile, long-term survival studies were performed using the slow-growing Dunning H rat prostate cancer model. For these latter studies, the dosing regimen was 10 mg CEP-701/kg/dose twice a day via gavage 5 days a week. This regimen maintains CEP-701 tumor tissue concentrations of 25-50 nM. Such chronic dosing increased (P < 0.001) the median survival of rats bearing the slow growing H prostate cancers from 408 days (395-432 days, 95% confidence interval) for the vehicle group (n = 18) to 566 days (497-598 days, 95% confidence interval) for the CEP-701-treated group (n = 24).


Subject(s)
Apoptosis/drug effects , Indoles , Neoplasm Metastasis/prevention & control , Prostatic Neoplasms/drug therapy , Receptor, trkA/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Cell Division/drug effects , Cell Line , Disease Models, Animal , Furans , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis/pathology , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/drug effects , Prostatic Neoplasms/pathology , Rats , Survival Analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Prostate ; 45(2): 140-8, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11027413

ABSTRACT

BACKGROUND: Prostatic cancer cells are lethal because they acquire the ability to activate survival pathways that do not require androgenic stimulation. As a rational approach to developing effective therapy for these devastating cells, specific signal transduction pathways uniquely required for the survival of these nonandrogen-dependent prostate cancer cells must be identified. Previous studies suggested that the neurotrophin/trk signal transduction axis may regulate such unique survival pathways. In the present study, the changes in expression of the neurotrophins (NGF, BDNF, and NT-3) and their cognate receptors (i.e., trk and p75NTR) during the progression of normal prostatic epithelial cells to malignancy were documented. Additionally, the consequences of inhibiting these trk signaling pathways on the in vitro survival of prostate cancer cells was tested. METHODS: Immmunocytochemistry, RT-PCR, and ELISA assays were used to characterize the changes in the neurotrophin ligands (i.e., NGF, BDNF, and NT-3) and their cognate high-affinity (i.e., trk A, B, and C) and low-affinity neurotrophin (i.e., p75 NTR) receptors in normal vs. malignant human prostatic tissues. CEP-751 is an indolocarbazole compound specifically designed to inhibit the initiation of these neurotrophin/trk signal transductions. The consequence of CEP-751 inhibition of trk signaling for in vitro clonogenic survival of a series of human prostatic cancer lines was also tested. RESULTS: These studies demonstrated that normal prostatic tissue from patients without prostate cancer contains substantial levels of nerve growth factor (NGF), which is produced in a paracrine manner by stromal cells. These stromal cells lack both trk and p75NTR receptors. In contrast, normal prostatic epithelial cells from patients without prostate cancer do not secrete detectable levels of neurotrophins, but do express trk A and p75 NTR. While the NGF/trkA/p75 NTR axis is present in the normal prostate, normal prostatic epithelial cells do not depend on this axis for their survival. In contrast, malignant prostate epithelial cells directly secrete a series of neurotrophins (i.e., NGF, BDNF, and/or NT-3) and express at least one if not more of the trk receptor proteins (i.e., trk A, B, and/or C), while no longer expressing the p75NTR receptors. In addition, inhibition of autocrine trk signaling via CEP-751 treatment induces the apoptotic death of these malignant cells. CONCLUSIONS: Prostate carcinogenesis involves molecular changes leading to the paracrine and/or autocrine production of a series of neurotrophins. This is coupled to the ectopic expression of trk B and trk C, as well as to the continued expression of trk A, and the loss of expression of p75NTR receptors. These changes result in the acquisition by malignant prostate cells of a unique requirement for trk signaling pathways for survival. Based on these findings, trk inhibition is a novel, rational approach for prostate cancer therapy.


Subject(s)
Nerve Growth Factors/metabolism , Prostatic Neoplasms/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Receptor, trkC/metabolism , Animals , Culture Media, Serum-Free , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Male , Mice , Nerve Growth Factors/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptor, Nerve Growth Factor/genetics , Receptor, trkA/genetics , Receptor, trkB/genetics , Receptor, trkC/genetics , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , Tumor Cells, Cultured
9.
Cancer Res ; 60(16): 4629-37, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969817

ABSTRACT

Eicosanoids modulate the interaction of tumor cells with various host components in cancer metastasis. Their synthesis involves the release of arachidonic acid (AA) from cellular phospholipids by phospholipase A2 (PLA2), followed by metabolism by cyclooxygenases (COXs) and lipooxygenases (LOXs). This study aimed to identify the pathway(s) of AA metabolism that are required for the invasion of prostate tumor cells. DU-145 and PC-3 human prostate cancer cell lines were used to test the effect of inhibitors of PLA2, COX, or LOX on the invasion of prostate tumor cells through Matrigel in vitro using the Boyden chamber assay and fibroblast-conditioned medium as the chemoattractant. We used nontoxic doses that did not inhibit simple cell motility and did not decrease clonogenic survival. All of the inhibitors caused a significant reduction in AA release from treated cells compared with control cells, which indicated that the treatments were biochemically active. Invasion through Matrigel was inhibited by the PLA2 inhibitor 4-bromophenacyl bromide (4-BPB), the general COX inhibitor ibuprofen (IB), and the highly selective COX-2 inhibitor NS398. Inhibition of cell invasiveness by 4-BPB (1.0 microM), IB (10.0 microM), and NS398 (10.0 microM) was reversed by the addition of prostaglandin E2 (PGE2). PGE2 alone, however, did not stimulate invasiveness, which suggests that its production is necessary for rendering the cells invasive-permissive but not sufficient for inducing invasiveness. In contrast, we found no significant inhibition of invasion of prostate tumor cells treated with esculetin (1.0 microM) or nordihydroguiaretic acid (1.0 microM), which are specific inhibitors of LOX. We also tested the effect of 4-BPB, IB, NS398, and esculetin on the secretion of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs), as key enzymes in the proteolysis of Matrigel during invasion, using gelatin zymograms and Western blots. Cells that received 4-BPB, IB, or NS398, but not esculetin showed a significant reduction in the levels of proMMP-2, MMP-9, and proMMP-9 in the culture medium. DU-145 cells did not secrete TIMP-1, and the drugs did not alter the secretion of TIMP-2. This work highlights the role played by COX in disturbing the balance between MMPs and TIMPs in prostate cancer cells, and it points to the potential use of COX inibitors, especially COX-2 selective inhibitors, in the prevention and therapy of prostate cancer invasion.


Subject(s)
Eicosanoids/physiology , Enzyme Inhibitors/pharmacology , Matrix Metalloproteinases/metabolism , Prostaglandins/biosynthesis , Prostatic Neoplasms/pathology , 3T3 Cells , Acetophenones/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arachidonic Acid/metabolism , Arachidonic Acid/physiology , Cell Movement/drug effects , Cell Survival/drug effects , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Eicosanoids/metabolism , Humans , Ibuprofen/pharmacology , Isoenzymes/antagonists & inhibitors , Lipoxygenase/metabolism , Lipoxygenase Inhibitors/pharmacology , Male , Matrix Metalloproteinase Inhibitors , Membrane Proteins , Mice , Neoplasm Invasiveness , Nitrobenzenes/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Prostaglandin-Endoperoxide Synthases , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Sulfonamides/pharmacology , Tissue Inhibitor of Metalloproteinases/metabolism , Tumor Cells, Cultured , Umbelliferones/pharmacology
10.
Prostate ; 43(4): 303-17, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10861750

ABSTRACT

BACKGROUND: New agents are required for the treatment of androgen-independent prostate cancer. Due to the low rate of proliferation of these malignant cells, agents which can activate the apoptotic death of these cells without requiring the cells being in the proliferative cell cycle are critically required. Thapsigargin (TG), via its ability to perturb intracellular free calcium [Ca(2+)](i), is such a cell proliferation-independent cytotoxic agent. The present study focuses on more completely describing the biochemical cascade during the apoptotic death of androgen-independent prostate cancer cells induced by TG and on the mechanistic requirements for this death. METHODS: A variety of cell and molecular biology techniques (e.g., time-lapse video, fluorescence image analysis, Northern and Western blotting) were used to examine the temporal relationship between changes in [Ca(2+)](i), GADD 153 transcription, translocation of the NFATc transcription factor to the nucleus, translocation of BAD from the cytosol to the mitochondria, caspase 9 activation, DNA fragmentation, and the loss of clonogenic survival induced by TG treatment of both human TSU-prl and rat AT3.1 prostate cancer cells in vitro. Additional studies using both microinjection of inhibitors of calmodulin and DNA transfections to induce expression of Ca(2+) binding proteins, e.g., calbindin, were performed to evaluate the causal relationship between [Ca(2+)](i) elevation, calmodulin/calcineurin activation, and apoptosis of prostate cancer cells. RESULTS: Using simultaneous fluorescence ratiometric and phase contrast image analysis in individual cells followed longitudinally for several days, it was documented that TG induced early (1-12 hr) moderate (i.e., <500 nM) elevation in [Ca(2+)](i). During this early rise in [Ca(2+)](i), genes like GADD 153 are induced at the transcriptional level. This early rise is followed by a return of [Ca(2+)](i) to baseline (i.e., approximately 50 nM) before the induction of a delayed (i.e., >12 hr) secondary rise ( approximately 10 microM) in [Ca(2+)](i). During the secondary rise in [Ca(2+)](i), Ca(2+) binds to calcineurin and calmodulin, allowing these proteins to form a complex which activates calcineurin's latent phosphatase activity. Once activated, calcineurin dephosphorylates NFATc and BAD, allowing translocation of these proteins to the nucleus and mitochondria, respectively. BAD translocation induces the release of cytochrome C from the mitochondria into the cytoplasm, which results in activation of caspase 9 and DNA fragmentation. If the TG-induced rise in [Ca(2+)](i) is blocked by overexpressing calbindin, or if calmodulin function is inhibited, these apoptotic events are prevented. CONCLUSIONS: TG induces the apoptotic death of prostate cancer cells via the activation of a reversible signaling phase induced by a transient nanomolar rise in [Ca(2+)](i), which involves new gene transcription and translation. This reversible signaling phase is followed by an irreversible commitment to undergo the execution phase which is induced by a secondary micromolar rise in [Ca(2+)](i). This secondary [Ca(2+)](i) rise irreversibly commits the cell to a calmodulin/calcineurin-dependent cascade, which results in DNA and cellular fragmentation into apoptotic bodies.


Subject(s)
Apoptosis , Calcineurin/physiology , Calmodulin/physiology , Enzyme Inhibitors/therapeutic use , Nuclear Proteins , Prostatic Neoplasms/pathology , Thapsigargin/therapeutic use , Animals , Calcium/metabolism , Carrier Proteins/metabolism , Cytochrome c Group/metabolism , DNA-Binding Proteins/metabolism , Humans , Male , NFATC Transcription Factors , Rats , Signal Transduction , Transcription Factors/metabolism , Tumor Cells, Cultured , bcl-Associated Death Protein
11.
Mol Hum Reprod ; 3(7): 563-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9268133

ABSTRACT

In order to test the hypothesis that integrin and uteroglobin (UG) expression in cultured endometrial cells are affected by hormone treatment, Ishikawa-CH endometrial cancer cells were cultured and exposed to oestradiol or oestradiol and progesterone regimens and assayed using immunohistochemistry. We evaluated the intensity of immunohistochemical staining for the integrin monomers alpha(v) and beta1, the dimers alpha(v)beta3 and alpha(v)beta6, and for the secretory protein uteroglobin under various experimental conditions. Cells grown in control media stained positively for the integrin monomers alpha(v) and beta1, the dimer alpha(v)beta3, and for UG. Oestradiol and sequential oestradiol/progesterone reversibly suppressed staining for the dimer alpha(v)beta3. Hormone treatment had no effect on the staining of the beta1 and alpha(v) monomers or UG. The alpha(v)beta6 dimer antibody did not stain under any experimental treatment conditions. These data indicate that expression of the integrin complex alpha(v)beta3 is reversibly suppressed by oestradiol in Ishikawa cells and that these cells may be a good model for studying hormone-driven molecular changes in endometrium.


Subject(s)
Endometrium/metabolism , Estradiol/pharmacology , Integrins/biosynthesis , Progesterone/pharmacology , Uteroglobin/biosynthesis , Adenocarcinoma , Embryo Implantation/physiology , Endometrial Neoplasms , Female , Humans , Tumor Cells, Cultured
12.
Clin Cancer Res ; 3(12 Pt 1): 2295-300, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9815627

ABSTRACT

We have shown previously that the secretory protein uteroglobin (UG) is highly expressed in normal human prostate tissue but this expression is either lost or altered in human prostate cancer cell lines. Treatment of these cell lines with recombinant human UG inhibits their ability to invade human reconstituted basement membrane by up to 90%, implying that the loss of normal UG expression may be related to the invasive potential of prostate cancer. Because invasion represents a critical step in metastasis, the expression patterns of UG could provide a unique and relevant indicator of cancer progression. In this study, we present the immunohistochemical analyses of fresh frozen prostate tissues from surgical specimens taken from 50 patients. Eight slides per patient were analyzed for UG staining. Slides from 26 patients showed evidence of prostate cancer, whereas slides from the remaining 24 patients showed only benign glands. The results demonstrate UG immunoreactivity in normal prostate, benign prostatic hyperplasia, and prostatic atrophy; low but clearly positive expression in prostatic intraepithelial neoplasia; positive expression in cancerous glands of Gleason's pattern

Subject(s)
Prostatic Neoplasms/pathology , Uteroglobin/analysis , Adult , Aged , Atrophy , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/surgery , Humans , Immunohistochemistry , Male , Middle Aged , Prostate/pathology , Prostatectomy , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/surgery , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery , Uteroglobin/genetics
13.
J Biol Chem ; 267(21): 15092-6, 1992 Jul 25.
Article in English | MEDLINE | ID: mdl-1378845

ABSTRACT

In addition to being regulated by a complex array of cis- and trans-acting factors, c-myc protooncogene expression may be modulated by antisense RNA transcripts. Our previous studies have determined that depletion of intracellular polyamines by alpha-difluoromethylornithine results in a marked decrease in the transcription of the human c-myc gene. Because of reports that antisense transcription occurs in the 5' and 3' regions of this gene, we used a genomic clone of the human c-myc gene to ascertain whether polyamine depletion might induce an antisense RNA transcript. These studies demonstrate that polyamine depletion of the human colon cancer cell line COLO 320 results in induction of an endogenous RNA transcript with high homology to the antisense strand of the second intervening sequence (PvuII-RsaI) of the c-myc gene. Furthermore, during such depletion, steady state levels of this transcript vary inversely to the sense direction c-myc RNA. RNase protection studies suggest that the antisense transcript may arise from a different gene locus than the c-myc gene. To further identify the origins of this RNA, a cDNA library was generated from size-selected RNA and screened with c-myc sequences. A 438-base pair cDNA was isolated with approximately 85% homology, to a 285-base region in the second intron of the c-myc gene. Computer homology analysis further reveals that a 120-base region within this cDNA also has approximately 85% homology to the antisense strands of a number of genes, including the growth-related genes, N-myc, p53, and thymidine kinase. These studies provide the initial characterization of an endogenous antisense RNA transcript which could influence cell growth by modulating the expression of c-myc and other genes.


Subject(s)
Genes, myc , RNA, Antisense/genetics , RNA/genetics , Transcription, Genetic , Base Sequence , Blotting, Northern , Colonic Neoplasms/metabolism , DNA , Humans , Molecular Sequence Data , Plasmids , RNA/metabolism , Restriction Mapping , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...