Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
J Clin Pathol ; 56(2): 107-13, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12560388

ABSTRACT

AIMS: To investigate the prognostic relevance of vascular endothelial growth factor (VEGF) and its receptor Flt-1 in nephroblastoma and whether tumour microvessel density (MVD) immunoreactivity, determined by the CD31 antigen, is related to the expression of VEGF and Flt-1. METHODS: The expression of VEGF and Flt-1 and MVD were investigated by means of immunohistochemical analysis in 62 Wilms's tumours. Patients were treated preoperatively with chemotherapy and had a mean follow up of 5.7 years. RESULTS: In general, VEGF and Flt-1 were expressed in normal kidney parenchyma and to a variable extent in the three main components of Wilms's tumour, namely: the blastemal, epithelial, and stromal cells. In tumour tissue, 52% and 47% of blastemal cells were positive for VEGF and Flt-1, respectively. A non-significant correlation was found between the expression of VEGF and Flt-1 in blastemal and epithelial cells and the clinicopathological stage. MVD was significantly higher in VEGF and Flt-1 positive tumours than in VEGF and Flt-1 negative tumours. Univariate analysis showed that the expression of VEGF and Flt-1 in blastemal cells was indicative of clinical progression and tumour specific survival. In addition, MVD expression was indicative of clinical progression. Epithelial staining was of no prognostic value. In a multivariate analysis, VEGF protein expression by blastemal cells was an independent prognostic marker for clinical progression. CONCLUSIONS: These results indicate that VEGF and Flt-1 protein expression are closely related to MVD and seem to be an important predictor for poor prognosis in treated patients with Wilms's tumour. Therefore, the expression of these molecules in primary Wilms's tumour may be useful in identifying those patients at high risk of tumour recurrence and in guiding antiangiogenic treatment.


Subject(s)
Kidney Neoplasms/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Wilms Tumor/metabolism , Analysis of Variance , Child , Child, Preschool , Disease Progression , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Kidney Neoplasms/blood supply , Kidney Neoplasms/therapy , Male , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Prognosis , Survival Rate , Wilms Tumor/blood supply , Wilms Tumor/therapy
2.
Cancer ; 92(12): 3120-9, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11753991

ABSTRACT

BACKGROUND: Wilms tumor is one of the most common solid tumors in children. A transforming growth factor-alpha (TGF-alpha)/epidermal growth factor receptor (EGF-R) autocrine loop plays an important role in tumor growth. Abnormal expression of TGF-alpha, EGF-R and c-erb B-2 has been demonstrated in several human malignancies. METHODS: The immunohistochemical expression of TGF-alpha, EGF-R, and c-erb B-2 was studied in paraffin material of 62 clinical Wilms tumors. Patients had a mean follow-up of 5.7 years. RESULTS: Generally, TGF-alpha, EGF-R, and c-erb B-2 were expressed in tissue of the normal kidney and at variable levels in the three cell types of Wilms tumor, i.e., blastemal, epithelial, and stromal cells. Immunoreactive blastema cells were found in 48%, 44%, and 34% of tumors for TGF-alpha, EGF-R, and c-erb B-2, respectively. It was found that TGF-alpha, EGF-R, and c-erb B-2 blastemal and epithelial expression gradually increased from T1 to T3. The blastemal expression of TGF-alpha was statistically significantly correlated with clinicopathologic stages. Both univariate and multivariate analysis showed that blastemal TGF-alpha expression was indicative for clinical progression, but neither blastemal TGF-alpha, nor EGF-R or c-erb B-2 expression correlated with patients survival. Epithelial staining was of no prognostic value. The simultaneous expression of TGF-alpha/EGF-R was indicative for clinical progression at univariate level. CONCLUSIONS: Increased expression of TGF-alpha in the blastemal part of Wilms tumor correlated with tumor classification and clinical progression. These findings suggest that significant expression of TGF-alpha and EGF-R may play a role in promoting transformation and/or proliferation of Wilms tumor, perhaps by an autocrine mechanism. Therefore, their expression may be of value in identifying patients at high risk of tumor recurrence.


Subject(s)
ErbB Receptors/biosynthesis , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Neoplasm Recurrence, Local , Receptor, ErbB-2/biosynthesis , Transforming Growth Factor beta/biosynthesis , Wilms Tumor/pathology , Biomarkers, Tumor/analysis , Child , Child, Preschool , Disease Progression , ErbB Receptors/analysis , Female , Humans , Immunohistochemistry , Kidney Neoplasms/genetics , Male , Prognosis , Receptor, ErbB-2/analysis , Risk Factors , Transforming Growth Factor beta/analysis , Wilms Tumor/genetics
3.
Br J Cancer ; 85(10): 1557-63, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11720445

ABSTRACT

Apoptotic cell death represents an important mechanism for the precise regulation of cell numbers in normal tissues. Various apoptosis-associated regulatory proteins, such as Bcl-2, Bax and Bcl-X, may contribute to the rate of apoptosis in neoplasia. The present study was performed to evaluate the prognostic value of these molecules in a group of 61 Wilms' tumours of chemotherapeutically pre-treated patients using an immunohistochemical approach. Generally, Bcl-2, Bax and for Bcl-X(S/L) were expressed in the blastemal and epithelial components of Wilms' tumour. Immunoreactive blastema cells were found in 53%, 41% and 38% of tumours for Bcl-2, Bax and for Bcl-X(S/L), respectively. An increased expression of Bcl-2 was observed in the blastemal component of increasing pathological stages. In contrast, a gradual decline of Bax expression was observed in the blastemal component of tumours with increasing pathological stages. Also blastemal Bcl-X(S/L) expression decreased with stage. Univariate analysis showed that blastemal Bcl-2 expression and the Bcl-2/Bax ratio were indicative for clinical progression, whereas epithelial staining was of no prognostic value. Multivariate analysis showed that blastemal Bcl-2 expression is an independent prognostic marker for clinical progression besides stage. These findings demonstrate that alterations of the Bcl-2/Bax balance may influence the clinical outcome of Wilms' tumour patients by deregulation of programmed cell death.


Subject(s)
Apoptosis , Biomarkers, Tumor/metabolism , Kidney Neoplasms/diagnosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Wilms Tumor/diagnosis , Child, Preschool , Disease Progression , Female , Humans , Immunohistochemistry , Kidney/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Prognosis , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , Treatment Outcome , Wilms Tumor/metabolism , Wilms Tumor/pathology , bcl-2-Associated X Protein , bcl-X Protein
4.
Int J Cancer ; 94(1): 28-34, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11668475

ABSTRACT

Most PCs show NE differentiation. Several studies have tried to correlate NE expression with disease status, but the reported findings have been contradictory. Prostatic NE cells synthesize peptides with a wide spectrum of potential functions. Some of these active peptides, such as PAMP, are amidated. PAM is the only carboxy-terminal peptide-amidating enzyme identified. We studied expression of PAMP and PAM in normal prostate and prostatic tumors (clinical specimens and human xenograft models) with or without prior androgen-deprivation therapy and found a wide distribution of both molecules in NE subpopulations of all kinds. Although the correlation of either marker to tumor grade, clinical progression or disease prognosis did not reach statistical significance, PAMP- or PAM-immunoreactive cells were induced after androgen-blockade therapy. In the PC-310 and PC-295 androgen-dependent models, PAMP or PAM NE differentiation was induced after castration in different ways, being higher in PC-310, which might explain its long-term survival after androgen deprivation. We show induction of expression of 2 new NE markers in clinical specimens and xenografted PC after endocrine therapy.


Subject(s)
Androgen Antagonists/therapeutic use , Mixed Function Oxygenases/analysis , Multienzyme Complexes/analysis , Neurosecretory Systems/cytology , Peptide Fragments/analysis , Peptides , Prostate/chemistry , Prostatic Neoplasms/chemistry , Proteins/analysis , Adrenomedullin , Animals , Cell Differentiation , Humans , Immunohistochemistry , Male , Mice , Neoplasm Transplantation , Prostatic Neoplasms/therapy , Transplantation, Heterologous
5.
Eur J Cancer ; 37(16): 2129-34, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11597395

ABSTRACT

Identification of genes involved in the transition from androgen-dependent to androgen-independent prostate cancer is important to extend our current knowledge of the disease. Using differential display RT-PCR analysis between androgen-dependent and androgen-independent prostate cancer cells, we have identified a novel gene, designated GC109. GC109 harbours a putative Cys-His cluster, a nuclear localisation signal, a leucine zipper and a ret finger protein (rfp)-like domain. GC109 mRNA expression in normal human tissues was found not to be restricted to the prostate. However, using a variety of 15 human cancer cell lines, GC109 mRNA was preferentially expressed in androgen-dependent LNCaP-FGC, compared with androgen-independent LNCaP-LNO, DU145 and PC3 human prostate cancer cells. Finally, the GC109 gene was mapped on human chromosome 2p24. Based on its protein domain structure and chromosomal localisation, we hypothesise that GC109 may be involved in chromosomal rearrangements in prostate cancer.


Subject(s)
Androgens/physiology , Chromosomes, Human, Pair 2/genetics , Gene Expression Regulation, Neoplastic , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Base Sequence , Blotting, Northern , Chromosome Mapping , Humans , Male , Molecular Sequence Data , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Anticancer Res ; 21(5): 3161-6, 2001.
Article in English | MEDLINE | ID: mdl-11848468

ABSTRACT

Renal Cell Carcinomas (RCCs) exhibit strong resistance to the most chemotherapeutic treatments probably due to the expression of various multidrug resistance (MDR) genes. Overexpression of P-glycoprotein (Pgp) is established as one such factor, but other mechanisms such as at-MDR, characterized by attenuated DNA-topoisomerase II (topoII) activity, may be functional as well. In addition, regulating proteins involved in apoptosis can exhibit multidrug resistant features. However, prevention of apoptosis as a mechanism of MDR has not yet been assessed in RCC, nor has the cytotoxicity of a variety of chemotherapeutic agents known to trigger apoptotic or necrotic cell death been tested in RCC in a systematic fashion. Using immunohistochemistry and Western blotting, Bcl-2 and Bax expression was determined in a panel of multidrug resistant RCC lines featuring Pgp and/or at-MDR. The results were related to apoptotic activity and kind of cell death in these cell lines, demonstrated by incubation with Hoechst 33342 and propidium iodide after treatment with various cytotoxic agents and quantitated by MTT. In the drug resistant sublines, some decreased Bax and strongly increased Bcl-2 expression was seen by immunohistochemistry indicating prevention of apoptosis as a distinct feature of MDR in RCC. This was confirmed by Western blotting. Sublines revealed significant resistance for all drugs, except for CC-313 and DiMIQ. However, these drugs induced necrotic cell death, in contrast to all other drugs tested, which induced apoptotic cell death. We conclude that, in chemoselected RCC sublines, multidrug resistance appears to be functional due to inhibition of apoptosis, apart from the MDR1 and at-MDR resistance mechanisms. CC-313 and DiMIQ are very potent cytotoxic agents in RCC, probably because they do not kill by induction of apoptosis.


Subject(s)
Apoptosis/drug effects , Carcinoma, Renal Cell/drug therapy , Drug Resistance, Multiple , Kidney Neoplasms/drug therapy , Amsacrine/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Topoisomerase II Inhibitors , Tumor Cells, Cultured , bcl-2-Associated X Protein
7.
Clin Cancer Res ; 6(11): 4265-71, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11106242

ABSTRACT

Wilms' tumor is one of the most common solid tumors of children. The protein product of the tumor-suppressor gene, Wilms' tumor 1 (WT-1), binds to the same DNA sequences as the protein product of the early growth response 1 (EGR-1) gene. There is experimental evidence that EGR-1 is involved in controlling cell growth. The expression of both genes in Wilms' tumor was studied by others, mainly at the mRNA level. The present study evaluates the prognostic value of WT-1 and EGR-1 in 61 Wilms' tumors of chemotherapeutically treated patients at the protein level, using an immunohistochemical approach. WT-1 was expressed in normal kidney tissues and in the blastemal and epithelial component of Wilms' tumor, whereas stromal tissue was negative. EGR-1 was expressed in normal kidney tissues and in the three main cell types of Wilms' tumor. In 59 and 56% of Wilms' tumor, the blastemal cells stained for WT-1 and EGR-1, respectively. The blastemal expression of WT-1 and EGR-1 and the epithelial expression of WT-1 were statistically significantly correlated with clinical stage. WT-1 immunoreactivity correlated with EGR-1 expression. Univariate analysis showed that blastemal WT-1 and EGR-1 expression were indicative for clinical progression and tumor-specific survival, whereas epithelial staining was of no prognostic value. Multivariate analysis showed that blastemal WT-1 expression is an independent prognostic marker for clinical progression other than stage. We conclude that a relationship exists between WT-1 and EGR-1 expression in clinical nephroblastomas. Blastemal WT-1 and EGR-1 expression is related to prognosis.


Subject(s)
DNA-Binding Proteins/analysis , Immediate-Early Proteins , Kidney Neoplasms/chemistry , Transcription Factors/analysis , Wilms Tumor/chemistry , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Humans , Immunoblotting , Immunohistochemistry , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Multivariate Analysis , Prognosis , Transcription Factors/genetics , WT1 Proteins , Wilms Tumor/mortality , Wilms Tumor/pathology
8.
J Natl Cancer Inst ; 92(17): 1414-21, 2000 Sep 06.
Article in English | MEDLINE | ID: mdl-10974077

ABSTRACT

BACKGROUND: The transition from androgen-dependent to androgen-independent prostate cancer is not fully understood but appears to involve multiple genetic changes. We have identified a gene, GC79, that is more highly expressed in androgen-dependent LNCaP-FGC human prostate cancer cells than in androgen-independent LNCaP-LNO human prostate cancer cells. Physiologic levels (0.1 nM:) of androgens repress expression of GC79 messenger RNA (mRNA) in LNCaP-FGC cells. To determine the role of GC79, we cloned its complementary DNA (cDNA) and functionally characterized its product. METHODS: The differentially expressed GC79 gene was cloned from human prostate cDNA libraries, sequenced, and transfected into mammalian cells to study its function. Expression of GC79 was analyzed in various adult and fetal human tissues and in prostate glands of castrated rats. The association of GC79 expression and apoptosis was investigated in COS-1 and LNCaP cells transfected with GC79 cDNA. All statistical tests are two-sided. RESULTS: Sequence analysis indicates that GC79 encodes a large, complex, multitype zinc-finger protein, containing nine C(2)H(2)-type zinc-finger domains, a cysteine-rich region, and a GATA C(4)-type zinc-finger domain. Castration-induced androgen withdrawal increased the expression of GC79 mRNA in the regressing rat ventral prostate, suggesting that the expression of GC79 mRNA is associated with the process of apoptotic cell death in the rat ventral prostate. Transfection and induction of GC79 cDNA in both COS-1 and LNCaP prostate cancer cells led to an apoptotic index that was eightfold higher (P:<.001, two-sided Student's t test) than that observed in uninduced transfected cells. CONCLUSIONS: We have cloned an androgen-repressible gene, GC79, that is potentially involved in apoptosis. This finding may have implications for the development of androgen-independent prostate cancer and, ultimately, for the treatment of prostate cancer.


Subject(s)
Androgens/metabolism , DNA-Binding Proteins , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Zinc Fingers/genetics , Amino Acid Sequence , Animals , Apoptosis , Base Sequence , Blotting, Northern , Cloning, Molecular , Gene Expression Regulation, Neoplastic , Humans , Male , Molecular Sequence Data , Prostatic Neoplasms/metabolism , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Repressor Proteins , Transcription Factors , Tumor Cells, Cultured
9.
Lab Invest ; 80(8): 1291-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10950120

ABSTRACT

Loss of the CD44 transmembrane glycoprotein in primary prostate cancer has been shown to be associated with unfavorable clinical behavior. Moreover, the majority of prostate cancer metastases lack expression of this molecule. The mechanism of CD44 silencing in prostate cancer was investigated using both patient material and in vivo-propagated human prostate cancer xenografts. In 9 of 11 lymph node metastases of prostate cancer, we demonstrated by methylation-sensitive restriction enzyme digestion that the promoter region of the CD44 gene is methylated, indicating that this represents a major mechanism of CD44 silencing. Similarly, in 6 out of 12 in vivo-growing human prostate carcinoma xenograft models, hypermethylation of the CD44 gene was found. The extent of CpG island methylation was investigated by nucleotide sequencing after bisulphite modification of the CD44 promoter region. In the xenografts displaying hypermethylation, the examined 14 CpG sites in the CD44 transcription regulatory domain, including a Sp1 binding site, were consistently methylated. This correlated with reduced CD44 expression or lack of CD44 expression at mRNA and protein levels. In the xenografts lacking hypermethylation of the CD44 gene, high levels of CD44 mRNA and protein were expressed in some models, whereas in others CD44 mRNA expression was only detectable by RT-PCR and the CD44 protein could hardly be detected or was not detected at all. The results indicate that, in most prostate cancers, loss of CD44 expression is associated with extensive hypermethylation of the CpG island of the CD44 promoter region, but other, posttranscriptional mechanisms may also lead to CD44 loss.


Subject(s)
DNA Methylation , Gene Silencing , Hyaluronan Receptors/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/immunology , Base Sequence , DNA Primers , Humans , Lymphatic Metastasis/immunology , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics
10.
Oncogene ; 19(6): 791-800, 2000 Feb 10.
Article in English | MEDLINE | ID: mdl-10698497

ABSTRACT

The Wilms' tumor 1 gene (WT1) encodes a transcription factor of the zinc-finger family and is homozygously mutated or deleted in a subset of Wilms' tumors. Through alternative mRNA splicing, the gene is expressed as four main polypeptides that differ by a stretch of 17 amino acids just N-terminal of the four zinc-fingers and three amino acids between zinc fingers 3 and 4. We have previously shown that expression of the WT1(-/-) isoform, lacking both inserts, increases the tumor growth rate of the adenovirus-transformed baby rat kidney (AdBRK) cell line 7C3H2, whereas expression of the WT1(-/+) isoform, lacking the 17aa insert, strongly suppresses the tumorigenic phenotype. In the present study we show that expression of these splice variants does not affect the tumorigenic potential of the similar AdBRK cell line, 7C1T1. In contrast to the 7C3H2 cell line, this AdBRK cell line expresses high endogenous levels of EGR-1 (early growth response-1) protein, a transcription factor structurally related to WT1. Ectopic expression of EGR-1 in the 7C3H2 AdBRK cells significantly increases their in vivo growth rate and nullifies the tumor suppressor activity of the WT1(-/+) protein. Furthermore, we find that EGR-1 levels are elevated in some Wilms' tumors. These data are the first to show that EGR-1 overexpression causes enhanced tumor growth and that WT1 and EGR-1 exert antagonizing effects on growth regulation in baby rat kidney cells, which might reflect the situation in some Wilms' tumors.


Subject(s)
DNA-Binding Proteins/physiology , Genes, Wilms Tumor , Immediate-Early Proteins , Neoplasm Proteins/physiology , Transcription Factors/physiology , Adenoviridae/physiology , Animals , Cell Division , Cell Line, Transformed , Cell Transformation, Viral , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Female , Gene Deletion , Kidney , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Transplantation , RNA Splicing , Rats , Transcription Factors/genetics , WT1 Proteins , Wilms Tumor/genetics , Wilms Tumor/pathology , Zinc Fingers/genetics
11.
Cancer Res ; 60(3): 741-8, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10676662

ABSTRACT

Neuroendocrine (NE) cells are androgen-independent cells and secrete growth-modulating neuropeptides via a regulated secretory pathway (RSP). We studied NE differentiation after androgen withdrawal in the androgen-dependent prostate cancer xenograft PC-310. Expression patterns of chromogranin A, secretogranin III, and prohormone convertase-1 were analyzed at both protein and mRNA level to mark the kinetics of NE differentiation both in vivo and in vitro. PC-310 tumor-bearing nude mice were killed at 0, 2, 5, 7, 14, and 21 days postcastration. PC-310C cultures initiated from collagenase-treated tumor tissue could be maintained up to four passages, and androgen-deprivation experiments were performed similarly. PC-310 tumor volumes decreased by 50% in 10 days postcastration. Proliferative activity and prostate-specific antigen (PSA) serum levels decreased to zero postcastration, whereas PSA levels in PC-310C culture media first decreased and subsequently increased after 5 days. In vivo, androgen receptor (AR) expression decreased initially but returned to control level from 5 days postcastration on. CgA, secretogranin III, and secretogranin V expression increased in vivo from 5 days postcastration on. Subsequently, prohormone convertase-1 and peptidyl alpha-amidating monooxygenase as well as the vascular endothelial growth factor were expressed from 7 days postcastration on, and, finally, growth factors such as gastrin-releasing peptide and serotonin were expressed in a small part of the NE cells 21 days postcastration. The PC-310 tumors did not show colocalization of the AR on the NE cells in the tumor residues after 21 days. As in the PC-310 xenograft, NE differentiation was induced and AR expression relapsed after prolonged androgen suppression in PC-310C. For PC-310C cells, this relapse was associated with the secretion of PSA. PC-310C is the first culture of human prostatic cancer cells having the NE phenotype. The PC-310 model system is a potential androgen-dependent model for studying the role of NE cells in the progression of clinical prostate cancer. Androgen deprivation of NE-differentiated prostate cancer may induce the formation of both NE- and AR-positive dormant tumor residues, capable of actively producing NE growth factors via a RSP, possibly leading to hormone refractory disease.


Subject(s)
Androgens/pharmacology , Multienzyme Complexes , Neoplasms, Hormone-Dependent/pathology , Neurosecretory Systems/cytology , Prostatic Neoplasms/pathology , Animals , Cell Differentiation , Chromogranin A , Chromogranins/analysis , Humans , Male , Mice , Mice, Nude , Mixed Function Oxygenases/analysis , Prostate-Specific Antigen/blood , Receptors, Androgen/analysis
12.
Br J Cancer ; 82(1): 112-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10638976

ABSTRACT

The possible role of activin in the regulation of malignant prostatic growth was studied using RNAase protection assays of activin receptors, inhibin/activin subunits and follistatin mRNAs in the human prostatic carcinoma cell lines LNCaP-FGC, -R and -LNO, in human prostatic carcinoma xenografts and in human prostatic tissue. Activin receptor types IA (ActRIA), IB (ActRIB), IIA (ActRIIA) and IIB (ActRIIB) mRNAs were generally expressed in prostate epithelial cells, with significantly lower levels of ActRIB mRNA in prostate tumour material when compared to non-malignant tissue (P < 0.05; Mann-Whitney U-test). Inhibin/activin betaA- and betaB-subunit mRNA expression was also found in prostate tissue. Androgen-independent xenografts expressed significantly lower amounts of betaB-subunit mRNA when compared to androgen-dependent xenografts (P< 0.05). While betaB-subunit mRNA was expressed by LNCaP-FGC and -LNO cells, virtually no expression was found in the androgen-independent LNCaP-R line. Inhibin alpha-subunit mRNA levels were low or undetectable in all samples investigated. Follistatin mRNA was undetectable in LNCaP-sublines, while low levels were found in prostatic tissues. In androgen-independent LNCaP-R cells, activin inhibited cell growth in a dose-dependent manner. These results suggest that prostate tumour progression is accompanied by a decrease of the inhibitory effect of locally produced activin by either a decrease in the expression of activin betaB-subunit mRNA or by a decrease of ActRIB mRNA levels.


Subject(s)
Glycoproteins/metabolism , Inhibins , Neoplasm Proteins/metabolism , Peptides/metabolism , Prostatic Neoplasms/metabolism , Prostatic Secretory Proteins , Receptors, Growth Factor/metabolism , Activin Receptors, Type I , Activin Receptors, Type II , Follistatin , Humans , Male , Prostate/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Prostate ; 42(1): 34-44, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10579797

ABSTRACT

BACKGROUND: Androgen-independent growth leads to progressive prostate cancer after androgen-ablation therapy. This may be caused by altered specificity of the androgen receptor (AR), by ligand-independent stimulation of the AR, or by paracrine growth modulation by neuropeptides secreted by neuroendocrine (NE) cells. METHODS: We established and characterized the androgen-independent FGC-DCC from the androgen-dependent LNCaP fast growing colony (FGC) cell line. The androgen-independent DU-145, FGC-DCC, and PC-3, and the androgen-dependent LNCaP and PC-346C cell lines were used to study growth modulation of gastrin-releasing peptide (GRP), calcitonin (CT), serotonin (5-HT), and vasoactive intestinal peptide (VIP) by (3)H-thymidine incorporation. Specificity of the growth-modulating effects was tested with the anti-GRP monoclonal antibody 2A11 and induction of cAMP by neuropeptides. RESULTS: Androgen-independent growth stimulation by neuropeptides was shown in DU-145 and PC-346C. 2A11 inhibited GRP-induced (3)H-thymidine incorporation in DU-145 and PC-346C and inhibited proliferation of the FGC-DCC and PC-3 cell lines. With some exceptions, cAMP induction paralleled growth stimulation. Dideoxyadenosine (DDA) inhibited the GRP-induced growth effect in DU-145 and PC-346C, whereas oxadiazoloquinoxaline-1-one (ODQ) had no effect on (3)H-thymidine incorporation. None of the neuropeptides stimulated growth of LNCaP, FGC-DCC, or PC-3. CONCLUSIONS: GRP-induced growth of DU-145 and PC-346C was specific and cAMP-mediated. Androgen-independent growth of FGC-DCC cells was mainly due to an induction of Bcl-2 expression and possibly through the activation of an autocrine and NE-like pathway, as has been shown also for the PC-3 cell line. Growth induction of non-NE cells by neuropeptides could be a possible role for NE cells in clinical prostate cancer.


Subject(s)
Androgens/physiology , Neuropeptides/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Cell Division/drug effects , Cyclic AMP/physiology , Dideoxyadenosine/pharmacology , Enzyme Inhibitors/pharmacology , Gastrin-Releasing Peptide/pharmacology , Humans , Male , Oxadiazoles/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinoxalines/pharmacology , Thymidine/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
14.
Int J Cancer ; 83(4): 506-11, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10508487

ABSTRACT

Using differential-display RT-PCR analysis between androgen-dependent LNCaP-FGC and androgen-independent LNCaP-LNO human prostate-cancer cells, we have identified a gene not previously described as being expressed in prostate. The gene is more highly expressed in androgen-independent than in androgen-dependent LNCaP prostate-cancer cells. Sequence analysis showed that the gene has already been cloned as a transcript present in embryonic brain, with unknown functions. Expression of the gene was found not to be restricted to the prostate, and not regulated by androgens in androgen-independent prostate-cancer cells. In concert with the cell-culture system, Northern-blot analysis of gene expression in vivo, using a panel of human prostate-cancer xenografts, demonstrated that the gene is more highly expressed in androgen-independent than in androgen-dependent prostate-cancer xenografts. The gene could be mapped on human chromosome 8q11. The 8q arm is known to be frequently amplified during prostate-cancer progression and harbors several proto-oncogenes potentially involved in cancer development. Since expression of the gene is positively correlated with prostate-cancer progression and its 8q11 chromosomal localization, we hypothesize that the gene may be involved in the development and progression of prostate cancer.


Subject(s)
Carcinoma/genetics , Chromosomes, Human, Pair 8/genetics , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Prostatic Neoplasms/genetics , Animals , Base Sequence , Blotting, Northern , Chromosome Mapping , Disease Progression , Humans , Male , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/genetics , RNA, Messenger/biosynthesis , Tumor Cells, Cultured
15.
Int J Cancer ; 84(5): 478-83, 1999 Oct 22.
Article in English | MEDLINE | ID: mdl-10502724

ABSTRACT

Decreased expression of CD44 is an independent prognostic marker for surgically treated prostate cancer. To investigate immunohistochemically defined CD44 expression in primary and metastatic prostate cancer, 2 groups of patients undergoing radical prostatectomy for clinically localized prostate cancer were studied. (1) pN1 group: 23 patients, finally staged pN1, of whom the radical prostatectomy specimen and the lymph nodes were investigated to establish a correlation between CD44 expression in the concurrently resected primaries and metastases; (2) pN(0) group: 23 patients with pN(0) disease matched for pT stage and Gleason sum score with the pN(1) patients. Progression rates based on serum prostate-specific antigen (PSA) levels could be determined in 42 of these 46 patients. In addition, 28 distant metastases were studied. A CD44 score of < 10% was found in 22 of the 23 lymph node metastases (96%) and in 20 of the corresponding radical prostatectomies. In the pN(0) group this was observed in only 6 out of 23 specimens. In most of the distant metastases CD44 scores were < 10%. Patients with pN0 disease and > 10% CD44-positive tumor cells had a significantly better prognosis than the other patients who were not significantly different from each other. CD44 expression is thus strongly reduced in prostate cancer metastases as well as in the corresponding primary tumors. This reduction may be used to predict the N stage clinically, provided that CD44 scores can be determined reliably on preoperative biopsy specimens.


Subject(s)
Hyaluronan Receptors/analysis , Prostatic Neoplasms/chemistry , Aged , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Prostatic Neoplasms/pathology
16.
Eur J Cancer ; 35(4): 664-71, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10492644

ABSTRACT

The aim of this study was to obtain insight into the role of the multidrug resistance (MDR) phenomenon in hormone-independent progressive prostate cancer. Using immunocytochemistry and Western blotting we determined the expression of P-glycoprotein (Pgp), multidrug resistance-associated protein (MRP), glutathione-S-transferase-pi (GST-pi), Bcl-2, Bax, topoisomerase (Topo) I, II alpha and II beta in the human prostate cancer cell lines PC3, TSU-Pr1, DU145 and LNCaP derivatives LNCaP-R, LNCaP-LNO and LNCaP-FGC. Proliferative activity was assessed by immunocytochemistry. MTT assays were used to determine the sensitivity to etoposide, doxorubicin and vinblastin. Pgp was not expressed in any of the cell lines. MRP was variably expressed. GST-pi was expressed in TSU-Pr1, PC3 and DU145. The expression of Bcl-2 was restricted to TSU-Pr1, whereas Bax was found in all cell lines. Topo II alpha was expressed at the highest level in the rapidly proliferating cell lines TSU-Pr1 and DU145. Topo I and II beta were equally expressed. Resistance profiles varied among the cell lines, with TSU-Pr1 being the most sensitive and LNCaP-LNO relatively resistant. Multiple MDR proteins were expressed in prostate cancer cell lines and may well influence response to chemotherapy. Future functional studies, using chemo-selected MDR models, may further help to determine the mechanism or combination of mechanisms underlying the resistance of prostate cancer to chemotherapy.


Subject(s)
Neoplasm Proteins/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blotting, Western , DNA Topoisomerases, Type I/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Glutathione Transferase/metabolism , Humans , Immunohistochemistry , Male , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured , bcl-2-Associated X Protein
17.
Br J Cancer ; 81(1): 28-36, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10487609

ABSTRACT

The beta 1-subunit of Na+,K+-ATPase was isolated and identified as an androgen down-regulated gene. Expression was observed at high levels in androgen-independent as compared to androgen-dependent (responsive) human prostate cancer cell lines and xenografts when grown in the presence of androgens. Down-regulation of the beta 1-subunit was initiated at concentrations between 0.01 nM and 0.03 nM of the synthetic androgen R1881 after relatively long incubation times (> 24 h). Using polyclonal antibodies, the concentration of beta 1-subunit protein, but not of the alpha 1-subunit protein, was markedly reduced in androgen-dependent human prostate cancer cells (LNCaP-FGC) cultured in the presence of androgens. In line with these observations it was found that the protein expression of total Na+,K+-ATPase in the membrane (measured by 3H-ouabain binding) was also markedly decreased. The main function of Na+,K+-ATPase is to maintain sodium and potassium homeostasis in animal cells. The resulting electrochemical gradient is facilitative for transport of several compounds over the cell membrane (for example cisplatin, a chemotherapeutic agent experimentally used in the treatment of hormone-refractory prostate cancer). Here we observed that a ouabain-induced decrease of Na+,K+-ATPase activity in LNCaP-FGC cells results in reduced sensitivity of these cells to cisplatin-treatment. Surprisingly, androgen-induced decrease of Na+,K+-ATPase expression, did not result in significant protection against the chemotherapeutic agent.


Subject(s)
Androgens/physiology , Neoplasms, Hormone-Dependent/enzymology , Prostatic Neoplasms/enzymology , Sodium-Potassium-Exchanging ATPase/biosynthesis , Animals , Antineoplastic Agents/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cisplatin/antagonists & inhibitors , Cisplatin/pharmacology , Down-Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Macromolecular Substances , Male , Metribolone/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Polymerase Chain Reaction , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Testosterone Congeners/pharmacology , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Urol Res ; 27(1): 3-8, 1999.
Article in English | MEDLINE | ID: mdl-10092147

ABSTRACT

In two androgen-dependent (FGC and P70) and two androgen-independent (LNO and R) sublines of the prostate cancer model LNCaP numerical and structural aberrations of chromosome 8 were investigated in detail. The techniques used were whole chromosome paint (WCP) and fluorescence in situ hybridization (FISH) with three cosmid probes mapping to different parts of the p-arm (D8S7 (8p23.3), LPL (8p22) and PLAT (8p11.1)). By WCP all four cell lines showed four copies of chromosome 8 in most cells. However, FISH demonstrated that in all sublines deletions in the 8p region were present. The majority of both FGC and P70 had two copies of cosmids D8S7 and LPL. The cosmid PLAT showed a broader distribution (1-4 copies), especially in P70. Compared with FGC and P70, both LNO and R showed a larger number of copies (3 or 4) of all three cosmid loci. It is discussed that this difference is probably the result of nondisjunction as a reaction to loss of other sequences on 8p, possibly the tumor suppressor gene (TSG) mapping to 8p21. The fact that both sublines LNO and R are androgen-independent raises the possibility of a link between TSG loss on 8p and androgen independence.


Subject(s)
Chromosome Aberrations/genetics , Chromosomes, Human, Pair 8/genetics , Prostatic Neoplasms/genetics , Cosmids/genetics , Gene Deletion , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Male , Molecular Biology/methods , Tumor Cells, Cultured
19.
Int J Cancer ; 80(3): 439-43, 1999 Jan 29.
Article in English | MEDLINE | ID: mdl-9935187

ABSTRACT

Down-regulation of the cell-surface adhesion molecule CD44 has been suggested to play an important role in tumor progression and metastasis of prostate cancer. CD44 is encoded by a gene that contains a CpG-rich region (CpG island) in its 5' regulatory sequence. We tried to assess whether hypermethylation of this region is the mechanism responsible for CD44 transcriptional inactivation. A panel of prostatic-carcinoma cell lines, Du145, LNCaP, PC3, PC346C and TSU, was analyzed for CD44 mRNA and protein expression. Du145, PC3 and TSU were positive for CD44, whereas in LNCaP and PC346C both CD44 mRNA and protein expression was suppressed. Methylation-sensitive restriction-enzyme analysis of genomic DNA showed that, in contrast to the CD44-positive cell lines, the CD44-negative lines were hypermethylated in the CD44 promoter CpG island. Furthermore, treatment of a PC346C culture with the demethylating agent 5-azacytidine resulted in re-expression of CD44 mRNA. It is concluded that hypermethylation of the CD44 5' promoter region is one of the mechanisms by which CD44 expression is down-regulated in prostatic-carcinoma cell lines.


Subject(s)
CpG Islands/physiology , DNA Methylation , DNA, Neoplasm/metabolism , Hyaluronan Receptors/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Down-Regulation , Humans , Hyaluronan Receptors/genetics , Male , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured
20.
Am J Pathol ; 154(2): 543-51, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10027412

ABSTRACT

It was previously shown in the PC-295 xenograft that the number of chromogranin A (CgA)-positive neuroendocrine (NE) cells increased after androgen withdrawal. NE cells did not proliferate and differentiated from G0-phase-arrested cells. Here we further characterized NE differentiation, androgen receptor status, and apoptosis-associated Bcl-2 expression in the PC-295 model after androgen withdrawal to assess the origin of NE cells. PC-295 tumor volumes decreased by 50% in 4 days. Intraperitoneal bromodeoxyuridine (BrdU) incorporation and MIB-1 labeling decreased to 0%, and the apoptosis was maximal at day 4. Androgen receptor expression and prostate-specific antigen (PSA) serum levels decreased rapidly within 2 days. The number of NE cells increased 6-fold at day 4 and 30-fold at day 7. Five and ten percent of the CgA-positive cells were BrdU positive after continuous BrdU labeling for 2 and 4 days, respectively. However, no MIB-1 expression was observed in CgA-positive cells. NE cells expressed the regulated secretory pathway marker secretogranin III but were negative for androgen receptor and Bcl-2. Bcl-2 expression did increase in the non-NE tumor cells. In conclusion, androgen withdrawal leads to a rapid PC-295 tumor regression and a proliferation-independent induction of NE differentiation. The strictly androgen-independent NE cells that were still present after 21 days differentiated mainly from G0-phase-arrested cells.


Subject(s)
Androgens/physiology , Neurosecretory Systems/pathology , Prostatic Neoplasms/pathology , Androgens/deficiency , Animals , Apoptosis , Cell Differentiation , Cell Division , Chromogranin A , Chromogranins/metabolism , Disease Models, Animal , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neurosecretory Systems/metabolism , Orchiectomy , Prostatic Neoplasms/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Androgen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...