Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Basic Res Cardiol ; 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796544

ABSTRACT

Multiple common cardiovascular comorbidities produce coronary microvascular dysfunction. We previously observed in swine that a combination of diabetes mellitus (DM), high fat diet (HFD) and chronic kidney disease (CKD) induced systemic inflammation, increased oxidative stress and produced coronary endothelial dysfunction, altering control of coronary microvascular tone via loss of NO bioavailability, which was associated with an increase in circulating endothelin (ET). In the present study, we tested the hypotheses that (1) ROS scavenging and (2) ETA+B-receptor blockade improve myocardial oxygen delivery in the same female swine model. Healthy female swine on normal pig chow served as controls (Normal). Five months after induction of DM (streptozotocin, 3 × 50 mg kg-1 i.v.), hypercholesterolemia (HFD) and CKD (renal embolization), swine were chronically instrumented and studied at rest and during exercise. Sustained hyperglycemia, hypercholesterolemia and renal dysfunction were accompanied by systemic inflammation and oxidative stress. In vivo ROS scavenging (TEMPOL + MPG) reduced myocardial oxygen delivery in DM + HFD + CKD swine, suggestive of a vasodilator influence of endogenous ROS, while it had no effect in Normal swine. In vitro wire myography revealed a vasodilator role for hydrogen peroxide (H2O2) in isolated small coronary artery segments from DM + HFD + CKD, but not Normal swine. Increased catalase activity and ceramide production in left ventricular myocardial tissue of DM + HFD + CKD swine further suggest that increased H2O2 acts as vasodilator ROS in the coronary microvasculature. Despite elevated ET-1 plasma levels in DM + HFD + CKD swine, ETA+B blockade did not affect myocardial oxygen delivery in Normal or DM + HFD + CKD swine. In conclusion, loss of NO bioavailability due to 5 months exposure to multiple comorbidities is partially compensated by increased H2O2-mediated coronary vasodilation.

3.
Biochem Pharmacol ; 93(3): 290-304, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25529535

ABSTRACT

Vemurafenib (PLX4032), an inhibitor of BRAF(V600E), has demonstrated significant clinical anti-melanoma effects. However, the majority of treated patients develop resistance, due to a variety of molecular mechanisms including MAPK reactivation through MEK. The induction of a cancer cell death modality associated with danger-signalling resulting in surface mobilization of crucial damage-associated-molecular-patterns (DAMPs), e.g. calreticulin (CRT) and heat shock protein-90 (HSP90), from dying cells, is emerging to be crucial for therapeutic success. Both cell death and danger-signalling are modulated by autophagy, a key adaptation mechanism stimulated during melanoma progression. However, whether melanoma cell death induced by MAPK inhibition is associated with danger-signalling, and the reliance of these mechanisms on autophagy, has not yet been scrutinized. Using a panel of isogenic PLX4032-sensitive and resistant melanoma cell lines we show that PLX4032-induced caspase-dependent cell death and DAMPs exposure in the drug-sensitive cells, but failed to do so in the drug-resistant cells, displaying heightened MEK activation. MEK inhibitor, U0126, treatment sensitized PLX4032-resistant cells to death and re-established their danger-signalling capacity. Only melanoma cells exposing death-induced danger-signals were phagocytosed and induced DC maturation. Although the PLX4032-resistant melanoma cells displayed higher basal and drug-induced autophagy, compromising autophagy, pharmacologically or by ATG5 knockdown, was insufficient to re-establish their PLX4032 sensitivity. Interestingly, autophagy abrogation was particularly efficacious in boosting cell death and ecto-CRT/ecto-HSP90 in PLX4032-resistant cells upon blockage of MEK hyper-activation by U0126. Thus combination of MEK inhibitors with autophagy blockers may represent a novel treatment regime to increase both cell death and danger-signalling in Vemurafenib-resistant metastatic melanoma.


Subject(s)
Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma , Signal Transduction/drug effects , Sulfonamides/pharmacology , Autophagy/physiology , Butadienes/pharmacology , Cell Death/drug effects , Cell Death/physiology , Coculture Techniques , Drug Resistance, Neoplasm/physiology , Enzyme Inhibitors/pharmacology , Humans , Indoles/therapeutic use , MAP Kinase Kinase Kinases/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Nitriles/pharmacology , Signal Transduction/physiology , Sulfonamides/therapeutic use , Vemurafenib
4.
Int J Androl ; 33(3): 507-17, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-19392831

ABSTRACT

Evidence from several models of hormone depletion and/or replacement and from knockout animals points to a key role of androgens in the control of spermatogenesis. In testes of mice with a Sertoli cell-selective ablation of the androgen receptor (SCARKO), transcriptional profiling, using microarray technology, revealed that, already on postnatal day 10,692 genes are differentially expressed compared with testes of control mice. Further evaluation of a subset of these genes by quantitative RT-PCR suggested that differences in expression may already be evident on day 8 or earlier. As the androgen receptor in mouse Sertoli cells becomes immunologically detectable around day 5, we tried to identify the earliest responses to androgens by a new transcriptional profiling study on testes from 6-day-old SCARKO and control mice. No obvious and novel early androgen response genes, potentially acting as mediators of subsequent indirect androgen actions, could be identified. However, several genes differentially expressed on day 10 already displayed a response to androgen receptor ablation on day 6. Quantitative RT-PCR studies for 12 of these genes on 10 paired SCARKO and control testes from 4-, 6-, 8-, 10-, 20- and 50-day-old mice revealed significant differences in expression level from day 4 onwards for three genes (Eppin, PCI, Cldn11) and from day 6 onwards for one more gene (Rhox5). For at least two of these genes (Rhox5 and Eppin), there is evidence for direct regulation via the androgen receptor. For three additional genes (Gpd1, Tubb3 and Tpd52l1) significantly lower expression in the SCARKO was noted from day 8 onwards. For all the studied genes, an impressive increase in transcript levels was observed between day 4-50 and differential expression was maintained in adulthood. It is concluded that the SCARKO model indicates incipient androgen action in mouse Sertoli cells from day 4 onwards.


Subject(s)
Receptors, Androgen/genetics , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Testis/metabolism , Androgens/genetics , Androgens/metabolism , Androgens/pharmacology , Animals , Crosses, Genetic , Female , Gene Expression/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Androgen/metabolism , Receptors, Androgen/physiology , Reverse Transcriptase Polymerase Chain Reaction , Spermatogenesis/drug effects , Spermatogenesis/genetics , Spermatogenesis/physiology , Testis/drug effects
5.
Methods ; 48(2): 193-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19362147

ABSTRACT

Prostate carcinoma is the most common non-cutaneous malignancy in males. Imaging of prostatic lesions is of great importance and aids in oncologic management and monitoring of therapy response. Particularly molecular imaging based on positron emission tomography (PET) and single photon emission computerized tomography (SPECT) has great potential. Using radio-labelled molecular probes, these approaches are highly sensitive and can provide key molecular and functional information on tumours. The identification of suitable targets based on unique genetic and biochemical features of cancer lesions is one of the core activities driving progress in molecular imaging of pathological processes. Nowadays, mainly metabolic probes are being used routinely for detection and staging of prostate cancer. The development of new specific receptor ligands and targeted probes and antibodies holds great promise to further enhance the performance of molecular imaging and to further improve the diagnosis and monitoring of prostate cancer.


Subject(s)
Molecular Diagnostic Techniques/methods , Prostatic Neoplasms/diagnosis , Animals , Antigens, Neoplasm , Bombesin , Cell Line, Tumor , Choline , Dideoxynucleosides , Dihydrotestosterone/analogs & derivatives , Fluorodeoxyglucose F18 , GPI-Linked Proteins , Humans , Male , Membrane Glycoproteins , Neoplasm Proteins , Positron-Emission Tomography/methods , Prostate/diagnostic imaging , Radiopharmaceuticals , Tomography, Emission-Computed, Single-Photon/methods
6.
J Mol Endocrinol ; 31(3): 583-96, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14664718

ABSTRACT

In search of potential androgen receptor coregulators we performed a yeast two-hybrid screening using the androgen receptor ligand-binding domain as bait and a human prostate cDNA library as prey and found that the carboxy-terminal domain of retinoblastoma-associated Krüppel protein (RbaK), a member of the Krüppel zinc finger protein family, interacts in a ligand-dependent way with the ligand-binding domain of the androgen receptor. RBaK was recently identified as a transcriptional regulator that interacts with the retinoblastoma protein and thereby influences E2F regulated transcription. The interaction of RBaK with the androgen receptor was further documented using mammalian two-hybrid experiments, in vitro binding studies and coimmunoprecipitation. Finally, we demonstrated that both RBaK and the retinoblastoma protein coactivate androgen receptor-mediated transcription in cotransfection experiments. In conclusion, our data show that RBaK interacts with the androgen receptor and increases its transcriptional activity. Moreover, the double interaction of RBaK with the retinoblastoma protein and with the androgen receptor provides a novel link between the androgen receptor and the regulation of the cell cycle.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Prostate/metabolism , Receptors, Androgen/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/physiology , Animals , COS Cells , Cell Cycle Proteins/genetics , Cells, Cultured , Chlorocebus aethiops , DNA-Binding Proteins/genetics , E2F Transcription Factors , Gene Library , Humans , Kruppel-Like Transcription Factors , Male , Receptors, Androgen/genetics , Retinoblastoma Protein/metabolism , Transcription Factors/genetics , Two-Hybrid System Techniques
7.
J Bone Miner Res ; 17(11): 2080-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12412816

ABSTRACT

This study was designed to evaluate the impact of estrogen versus androgen action on orchidectomy (ORX)-induced bone loss and associated changes in body composition. During an experimental period of 4 months, aged (12-month-old) ORX rats were treated with 17beta-estradiol (E2; 0.75 microg/day) or different doses of the nonaromatizable androgen 5alpha-dihydrotestosterone (DHT; 45, 75, and 150 microg/day, respectively), via subcutaneous (sc) silastic implants. Low doses of DHT and E2 inhibited the ORX-induced rise of bone turnover markers (serum osteocalcin and urinary deoxypyridinoline [DPD]) to a similar extent. High-dose DHT prevented the ORX-induced decrease of trabecular bone density but had no significant effect on cortical thinning as assessed by peripheral quantitative computed tomography (pQCT). This bone-sparing action of DHT occurred at the expense of hypertrophy of the ventral prostate and seminal vesicles. On the other hand, E2 restored both trabecular bone density and cortical thickness in ORX rats and even prevented age-related bone loss. In contrast to DHT, E2 increased lean body mass and inhibited the ORX-associated increase of fat mass, as measured by DXA. Administration of E2 was associated with increased serum concentrations of insulin-like growth factor (IGF) I and decreased circulating levels of leptin. We conclude that, in the aged ORX rat model, E2 is more effective in preventing ORX-induced bone loss than DHT. Additionally, E2 has anabolic effects on muscle tissue and prevents the ORX-related increase of fat mass. Overall, these data suggest that androgen action on bone and body composition is dependent on stimulation of both androgen receptors (ARs) and estrogen receptors (ERs).


Subject(s)
Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Osteoporosis/drug therapy , Aging/physiology , Anabolic Agents/pharmacology , Animals , Body Composition/drug effects , Bone Density/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Male , Orchiectomy , Rats
8.
Calcif Tissue Int ; 70(3): 170-5, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11907714

ABSTRACT

Testosterone (T) can affect bone metabolism not only directly, but also via its metabolites, estrogen or dihydrotestosterone, produced by enzymes present in bone. Therefore, the aim of this study was to investigate whether the high-affinity estrogen receptor ligand ICI 182,780 (ICI) impaired the bone-protective action of T in 3-month-old orchidectomized (Orch) rats, studied during an experimental period of 3 months. As expected, Orch significantly decreased trabecular bone volume in the proximal tibial metaphysis (-52%), as measured by histomorphometry, and had a similar negative effect on volumetric bone mineral density (BMD) in the distal femoral metaphysis (-53%), as assessed by peripheral quantitative computed tomography (pQCT). The loss of bone induced by Orch was completely prevented by T administration. Moreover, the Orch-associated increases of biochemical markers of bone turnover (serum osteocalcin, urinary deoxypyridinoline, and calcium excretion) did not occur when Orch rats received T. Administration of ICI in combination with T did not impair this bone-sparing effect. Cortical bone parameters (as determined by pQCT), body weight gain, and body composition (as measured by dual-energy X-ray absorptiometry) were not affected by T or ICI in combination with T. Furthermore, no differences were observed in serum concentrations of insulin-like growth factor-I or glucose homeostasis. In conclusion, ICI does not impair the long-term bone-protective effects of T in orchidectomized male rats, suggesting that testosterone can mediate its effect on the male skeleton directly via the androgen receptor. The absence of effects on body growth via the growth hormone--insulin-like growth factor-I axis may be a possible explanation for the lack of skeletal effects of this selective estrogen receptor antagonist.


Subject(s)
Bone and Bones/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Receptors, Estrogen/metabolism , Testosterone/pharmacology , Animals , Biomarkers/analysis , Bone and Bones/metabolism , Drug Interactions , Estradiol/metabolism , Estrogen Antagonists/pharmacology , Estrogens/metabolism , Follow-Up Studies , Fulvestrant , Ligands , Male , Models, Animal , Orchiectomy/adverse effects , Rats , Rats, Wistar , Receptors, Androgen/metabolism , Testosterone/metabolism
9.
Mol Endocrinol ; 15(10): 1817-28, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11579213

ABSTRACT

Using two independent prostate cancer cell lines (LNCaP and MDA-PCa-2a), we demonstrate that coordinated stimulation of lipogenic gene expression by androgens is a common phenomenon in androgen-responsive prostate tumor lines and involves activation of the sterol regulatory element-binding protein (SREBP) pathway. We show 1) that in both cell lines, androgens stimulate the expression of fatty acid synthase and hydroxymethylglutaryl-coenzyme A synthase, two key lipogenic genes representative for the fatty acid and the cholesterol synthesis pathway, respectively; 2) that treatment with androgens results in increased nuclear levels of active SREBP; 3) that the effects of androgens on promoter-reporter constructs derived from both lipogenic genes (fatty acid synthase and hydroxymethylglutaryl-coenzyme A synthase) depend on the presence of intact SREBP-binding sites; and 4) that cotransfection with dominant-negative forms of SREBPs abolishes the effects of androgens. Related to the mechanism underlying androgen activation of the SREBP pathway, we show that in addition to minor effects on SREBP precursor levels, androgens induce a major increase in the expression of sterol regulatory element-binding protein cleavage-activating protein (SCAP), an escort protein that transports SREBPs from their site of synthesis in the endoplasmic reticulum to their site of proteolytical activation in the Golgi. Both time course studies and overexpression experiments showing that increasing levels of SCAP enhance the production of mature SREBP and stimulate lipogenic gene expression support the contention that SCAP plays a pivotal role in the lipogenic effects of androgens in tumor cells.


Subject(s)
Androgens/pharmacology , CCAAT-Enhancer-Binding Proteins/genetics , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Lipids/biosynthesis , Membrane Proteins/genetics , Prostatic Neoplasms/metabolism , Transcription Factors , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/physiology , COS Cells , Cell Nucleus/metabolism , Cholesterol/biosynthesis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Fatty Acid Synthases/genetics , Fatty Acids/biosynthesis , Genes, Reporter , Humans , Hydroxymethylglutaryl-CoA Synthase/genetics , Intracellular Signaling Peptides and Proteins , Kinetics , Male , Membrane Proteins/physiology , Mutagenesis , Point Mutation , Promoter Regions, Genetic , Sterol Regulatory Element Binding Protein 1 , Structure-Activity Relationship , Transfection , Tumor Cells, Cultured
10.
Verh K Acad Geneeskd Belg ; 63(4): 321-33, 2001.
Article in English | MEDLINE | ID: mdl-11603058

ABSTRACT

Steroid hormones have a strong influence on the biology of several common human cancers, including cancer of the prostate, breast, endometrium and ovarium. To gain more insight into this process, a screening for androgen-regulated genes was set up in prostate cancer cells. In addition to their well known effects on cell survival, proliferation and differentiated function, androgens were found to markedly stimulate the expression of several lipogenic enzymes. In clinical cancer samples these enzymes are markedly overexpressed in comparison to normal tissues, allowing them to be used as cancer markers and as potential targets for antineoplastic therapy. Investigation of the underlying mechanisms of gene regulation revealed that androgens stimulate lipogenic gene expression through a novel indirect mechanism involving Sterol Regulatory Element-binding Proteins (SREBPs), lipogenic transcription factors that play a key role in the fundamental feedback mechanism of cellular lipid homeostasis. Interestingly, also growth factors, whose signaling pathways are frequently dysregulated and constitutively activated as prostate cancer progresses towards a more advanced disease, stimulate lipogenesis through the same SREBP-mediated mechanism. While studies on the role of enhanced intermediary metabolism in cancer cell biology are progressing, these findings provide important new insights into the long-known dysregulation of intermediary metabolism in cancer cells and open new perspectives for clinical diagnosis and therapeutic intervention.


Subject(s)
Androgens/metabolism , Gene Expression Regulation, Neoplastic , Lipids/biosynthesis , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Biomarkers , CCAAT-Enhancer-Binding Proteins/physiology , DNA-Binding Proteins/physiology , Humans , Lipid Metabolism , Lipids/genetics , Male , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Transcription Factors/physiology , Tumor Cells, Cultured
11.
Prostate ; 48(2): 104-17, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11433420

ABSTRACT

BACKGROUND: Prostatic stroma affects both proliferation and differentiation of epithelial cells but the factors involved remain poorly understood. In order to identify and characterize potential paracrine mediators, we studied the effects of human prostate fibroblast-conditioned media (PFCM) in three bioassay systems. METHODS: The first bioassay uses transferrin secretion by cultured rat Sertoli cells as an endpoint for differentiating activity. Factors active in this (heterologous) assay were compared to PModS, a mediator of mesenchymal-epithelial interactions in the testis, also produced by rat prostate stromal cells. The two other (homologous) bioassays use LNCaP tumor cells or subcultured human prostate epithelial cells (PEC) as targets. Differentiation is evaluated by prostate-specific antigen (PSA) secretion and reverse transcriptase-polymerase chain reaction (RT-PCR) for a number of markers of epithelial function. Proliferation is assayed by measurements of DNA and thymidine incorporation. RESULTS: PFCM markedly stimulates transferrin production by Sertoli cells. The main factor(s) involved are acid stable and bind to heparin. However, both their size (approximately 37 kDa) and their behavior on reversed-phase chromatography differ from that of PModS. Although PFCM increases total RNA content of LNCaP, it does not increase or restore differentiated function of LNCaP or PEC. Proliferative effects are observed in LNCaP and these effects cannot be neutralized by an antiserum directed against basic fibroblast growth factor (bFGF). Antiproliferative effects are observed in PEC and these effects are largely due to transforming growth factor-beta (TGF-beta). CONCLUSIONS: PFCM provokes differentiating effects in a Sertoli cell bioassay, but unlike with rat stromal cells, the factor(s) involved differ from PModS. In the two homologous systems studied, differentiating effects could not be demonstrated and discordant effects were noted on proliferation. Various bioassay systems will be required to identify the spectrum of mediators present in PFCM.


Subject(s)
Cell Differentiation , Paracrine Communication/physiology , Prostate/physiology , Sertoli Cells/physiology , Animals , Biological Assay , Cell Communication , Cell Culture Techniques , Cell Division , Culture Media , Humans , Male , Mesoderm/physiology , Prostate/cytology , Rats , Transferrin/biosynthesis , Transforming Growth Factor beta
12.
Biochem Biophys Res Commun ; 285(1): 70-6, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11437374

ABSTRACT

To examine the role of the estrogen receptor-alpha (ERalpha) during male skeletal development, bone density and structure of aged ERalphaKO mice and wild-type (WT) littermates were analyzed and skeletal changes in response to sex steroid deficiency and replacement were also studied. In comparison to WT, ERalphaKO mice had smaller and thinner bones, arguing for a direct role of ERalpha to obtain full skeletal size in male mice. However, male ERalphaKO mice had significantly more trabecular bone as assessed both by pQCT and histomorphometry, indicating that ERalpha is not essential to maintain cancellous bone mass. Six weeks following orchidectomy (ORX), both WT and ERalphaKO mice showed high-turnover osteoporosis as revealed by increases in serum osteocalcin and decreases in trabecular (-38% and -58% in WT and ERalphaKO, respectively) and cortical bone density (-5% and -4% in WT and ERalphaKO, respectively). Administration of testosterone propionate (T, 5 mg/kg/day) completely prevented bone loss both in ERalphaKO and in WT mice. As expected, estradiol (E2, 60 microg/kg/day) replacement did not prevent cancellous bone loss in ORX ERalphaKO mice. However, E2 stimulated bone formation at the endocortical surface in ORX ERalphaKO, suggesting that osteoblasts may respond to nonERalpha-mediated estrogen action. In conclusion, although functional ERalpha may play a significant role during male skeletal development, this receptor does not seem essential for androgen-mediated skeletal maintenance in older male mice.


Subject(s)
Orchiectomy/adverse effects , Osteoporosis/prevention & control , Receptors, Estrogen/physiology , Testosterone/pharmacology , Animals , Bone Density , Estrogen Receptor alpha , Male , Mice , Mice, Knockout , Osteoporosis/etiology , Receptors, Estrogen/genetics
13.
Biochem Biophys Res Commun ; 283(1): 97-101, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11322773

ABSTRACT

Androgens exert a peculiar biphasic dose-dependent influence on the proliferation of LNCaP cells, a widely used model to study androgen effects on prostate cancer cells. Low concentrations of androgen stimulate proliferation, but high concentrations inhibit proliferation and induce strong expression of differentiation markers. In order to gain more insight into the molecular mechanisms that underlie these changes we studied the influence of a wide concentration range of the synthetic androgen R1881 on several cell cycle- and differentiation-related parameters. Low concentrations (0.1 nM), known to promote LNCaP cell proliferation, induce an increase of Retinoblastoma protein phosphorylation, accompanied by an increase of E2F-1 protein levels and E2F activity and by increased expression of the E2F-target gene products E2F-1 and cyclin A. High concentrations of R1881 (10 nM) induce strong expression of the differentiation marker prostate-specific antigen. Retinoblastoma protein is largely hypophosphorylated, resulting in low E2F activity and low concentrations of E2F-1 and cyclin A mRNA. Finally, there is a strong increase of p27(KIP1) protein, but not of p27(KIP1) mRNA. These results indicate that the biphasic dose response of LNCaP proliferation to androgen is closely reflected in Rb phosphorylation, E2F activity and p27(KIP1) protein expression.


Subject(s)
Androgens/metabolism , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins , Androgens/pharmacology , Blotting, Western , Cell Division/drug effects , Cyclin A/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Dose-Response Relationship, Drug , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Inhibitors/metabolism , Genes, Reporter/drug effects , Humans , Male , Metribolone/pharmacology , Microtubule-Associated Proteins/metabolism , Phosphorylation/drug effects , RNA, Messenger/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transfection , Tumor Cells, Cultured
14.
Mol Cell Endocrinol ; 168(1-2): 21-9, 2000 Oct 25.
Article in English | MEDLINE | ID: mdl-11064149

ABSTRACT

Transient cotransfection in COS-7 cells, a standard approach to demonstrate coactivation, was used to study the coactivation properties of NuRIP183, a new nuclear receptor interacting protein of 183 kDa, isolated by a yeast two-hybrid screening. Transfection with a NuRIP183 expression construct strongly increased the ligand-dependent response of reporter constructs for several nuclear receptors when compared to transfection with the empty expression vector. A more detailed study, however, revealed major changes in the expression level of the nuclear receptors in cotransfection experiments, indicating that the observed changes in receptor activity were not due to coactivation but to differences in receptor concentration caused by interference from the cotransfected expression constructs with the expression of the receptor. Such interference, which is inversely related to the length of the insert, was observed, not only in COS-7 cells but also in CV-1 and MCF-7 cells, using different transfection techniques (FuGENE-6 and calcium phosphate) and different expression vectors (pSG5, pcDNA1.1 and pIRESneo). These data cast some doubt on coactivation of nuclear receptors based on similar cotransfection experiments without measurement of receptor concentration. Moreover, it is recommended to limit the amounts of (co)transfected expression plasmid and to avoid the use of empty expression plasmid as a control. Finally, one should be aware of similar misleading results in other experimental set-ups based on cotransfection.


Subject(s)
Gene Expression Regulation , Nuclear Proteins/metabolism , Receptors, Androgen/metabolism , Transfection/methods , Animals , Artifacts , Blotting, Northern , Blotting, Western , COS Cells , Gene Expression , Genes, Reporter , Genetic Vectors , Molecular Weight , Nuclear Proteins/chemistry , Receptors, Androgen/genetics , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Two-Hybrid System Techniques
15.
Oncogene ; 19(45): 5173-81, 2000 Oct 26.
Article in English | MEDLINE | ID: mdl-11064454

ABSTRACT

Increased expression of fatty acid synthase (FAS) is observed in a clinically aggressive subset of various common cancers and interference with FAS offers promising opportunities for selective chemotherapeutic intervention. The mechanisms by which FAS expression is (up)-regulated in these tumors remain, however, largely unknown. Recently we demonstrated that in LNCaP prostate cancer cells FAS expression is markedly elevated by androgens via an indirect pathway involving sterol regulatory element-binding proteins (SREBPs). Here, we also show that growth factors such as EGF are able to stimulate FAS mRNA, protein and activity. Several observations also indicate that the effects of EGF on FAS expression are ultimately mediated by SREBPs. EGF stimulates SREBP-1c mRNA expression and induces an increase in mature nuclear SREBP-1. Moreover, in transient transfection studies EGF stimulates the transcriptional activity of a 178 bp FAS promoter fragment harboring a complex SREBP-binding site. Deletion or mutation of this binding site abolishes these effects and ectopic expression of dominant negative SREBP-1 inhibits FAS expression and induction in intact LNCaP cells. Given the frequent dysregulation of growth factor signaling in cancer and the key role of SREBP-1 in lipid homeostasis, growth factor-induced activation of the SREBP pathway is proposed as one of the mechanisms responsible for up-regulation of lipogenic gene expression in a subset of cancer cells.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Epidermal Growth Factor/pharmacology , Fatty Acid Synthases/biosynthesis , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Transcription Factors , Binding Sites , CCAAT-Enhancer-Binding Proteins/genetics , Carcinoma , DNA-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic , Humans , Male , Promoter Regions, Genetic , Prostatic Neoplasms/enzymology , RNA, Messenger/biosynthesis , Regulatory Sequences, Nucleic Acid , Signal Transduction , Sterol Regulatory Element Binding Protein 1 , Tumor Cells, Cultured , Up-Regulation
16.
Int J Cancer ; 88(2): 176-9, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11004665

ABSTRACT

A substantial subset of breast, colorectal, ovarian, endometrial and prostatic cancers displays markedly elevated expression of immunohistochemically detectable fatty acid synthase, a feature that has been associated with poor prognosis and that may be exploited in anti-neoplastic therapy. Here, using an RNA array hybridisation technique complemented by in situ hybridisation, we report that in prostate cancer fatty acid synthase expression is up-regulated at the mRNA level together with other enzymes of the same metabolic pathway. Contrary to the observations that in many cell systems (including androgen-stimulated LNCaP prostate cancer cells) fatty acid and cholesterol metabolism are co-ordinately regulated so as to supply balanced amounts of lipids for membrane biosynthesis, storage or secretion, no changes in the expression of genes involved in cholesterol synthesis were found. These findings point to selective activation of the fatty acid synthesis pathway and suggest a shift in the balance of lipogenic gene expression in a subgroup of prostate cancers.


Subject(s)
Fatty Acid Synthases/genetics , Gene Expression Regulation, Neoplastic , Prostate/enzymology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Secretory Proteins , Transcription, Genetic , Acetyl-CoA Carboxylase/genetics , Acid Phosphatase/genetics , Gene Expression Regulation, Enzymologic , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , In Situ Hybridization , Male , Peptides/genetics , Prostatic Hyperplasia/enzymology , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , RNA, Messenger/genetics , Reference Values
17.
Endocrinology ; 141(5): 1642-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10803572

ABSTRACT

The aim of this study was to evaluate the effects of different doses of androgen replacement, both on body composition and bone, in an aged male orchidectomized rat model. Testosterone was administered by 0.5, 1, and 2.5-cm sc SILASTIC implants (release of, respectively, 11.5, 23, and 55 microg/day) to aged (12 months old, +/- 550 g) male orchidectomized Wistar rats during a 15-week experimental period. T 0.5 only partially prevented decrease of ventral prostate and seminal vesicle weight, compared with an intact group that received an empty implant (Intact). The 1-cm implant (T 1) completely prevented decrease of both seminal vesicles and ventral prostate weight. The 2.5-cm implant (T 2.5) was clearly supraphysiological, as demonstrated by significant hypertrophy of both androgen-sensitive organs. Serum testosterone was lower in T 0.5 and T 1 (0.38 +/- 0.06 ng/ml and 0.92 +/- 0.06 ng/ml, respectively) and higher in T 2.5 (2.4 +/- 0.28. ng/ml), compared with both Intact (1.6 +/- 0.23 ng/ml) and the baseline group(1.6 +/- 0.11 ng/ml). As expected, orchidectomized rats that received an empty SILASTIC implant had significantly lower bone mineral content (-7.9%), apparent density (-5.7%), and lean body mass (-10.8%), as measured by dual-energy x-ray absorptiometry, without significant changes in body weight and fat mass, compared with Intact. Also, cancellous (-50.3%) and cortical (-1.8%) volumetric density, as measured by peripheral quantitative computed tomography, were decreased in the tibia. Bone turnover, as measured by serum osteocalcin and urinary deoxypyridinoline excretion, was increased in orchidectomized rats that received an empty SILASTIC implant. T 0.5 prevented all changes, not only in bone mineral content, density, and turnover but also in lean body mass. Moreover, there were no significant differences, for all these parameters, between the different doses of testosterone replacement. In conclusion, low-dose androgen replacement does not lead to lower bone mineral density, higher bone turnover, and lower lean body mass in aged male rats, whereas complete androgen deficiency does. Therefore, the threshold concentration of testosterone necessary for prevention of both bone and lean body mass loss in aged male rats is clearly lower than for prostate and seminal vesicles.


Subject(s)
Aging/pathology , Androgens/physiology , Body Composition/drug effects , Bone Resorption/physiopathology , Androgens/administration & dosage , Androgens/therapeutic use , Animals , Body Mass Index , Bone Density , Disease Models, Animal , Male , Orchiectomy , Rats
18.
Biochem Biophys Res Commun ; 269(1): 209-12, 2000 Mar 05.
Article in English | MEDLINE | ID: mdl-10694501

ABSTRACT

Enhanced expression of fatty acid synthase and other lipogenic enzymes has been observed in a subset of breast cancers with poor prognosis. This phenomenon has been related to amplification of a gene on chromosome region 11q13 encoding Spot 14, a putative regulator of lipogenic enzyme expression. In this paper we demonstrate that the induction of lipogenesis by progestins and androgens in the breast cancer cell line T47-D is accompanied by a marked increase in the expression of Spot 14. These data corroborate the correlation between Spot 14 expression and increased lipogenesis. Moreover they show that apart from gene amplification there is another steroid-regulated pathway that may enhance Spot 14 expression and lipogenesis in tumor cells.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Progesterone Congeners/pharmacology , Proteins/genetics , Testosterone Congeners/pharmacology , Chromosomes, Human, Pair 11/genetics , Dihydrotestosterone/pharmacology , Female , Gene Expression/drug effects , Humans , Lipids/biosynthesis , Metribolone/pharmacology , Nuclear Proteins , Pregnenediones/pharmacology , Progesterone/pharmacology , Transcription Factors , Tumor Cells, Cultured
19.
Mol Cell Endocrinol ; 155(1-2): 9-18, 1999 Sep 10.
Article in English | MEDLINE | ID: mdl-10580834

ABSTRACT

Changes in circulating levels of insulin-like growth factors (IGF) and IGF-binding proteins (IGFBP) have been related to prostate cancer, but the nature and the significance of this relationship remains elusive. Recent reports suggest that modulation of the production of IGFBP-3 by retinoids may affect growth of breast and prostate tumor cells. In the present study we explored whether androgens (R1881), retinoids (all-trans- and 9-cis-retinoic acid: atRA and 9cRA), deltanoids (1alpha,25-dihydroxycholecalciferol: VD3) and thyroid hormone (triiodothyronine: T3) influence the production of IGFBPs by LNCaP prostatic adenocarcinoma cells and whether the observed changes affect tumor cell growth. Northern blot experiments demonstrated that LNCaP cells express IGFBP-2, -3, -4 and (to a small extent) -5. IGFBP-4 and -5 were not measurably affected by the mentioned agonists. At a growth promoting concentration (10(-10) M), R1881 increased IGFBP-2 transcript levels two- to three-fold and this effect was neutralized by atRA and VD3. Similar effects could not be demonstrated, however, at the protein level using Western ligand blotting. R1881 decreased and atRA increased the mRNA levels of IGFBP-3 and these effects were confirmed by Western ligand blotting and by radioimmunoassay. The effects of atRA were mimicked by 9cRA and by a specific RAR agonist but not by a RXR agonist. VD3 and T3 had no significant effect on IGFBP-3 secretion but respectively enhanced or decreased the effect of 9cRA. The effects of retinoids required high concentrations (10(-6)-10(-5) M) that also induced growth inhibition. R1881, however, decreased IGFBP-3 at growth promoting (10(-10) M) as well as at growth inhibitory (10(-8) M) concentrations. Moreover, under serum-free conditions, we were unable to demonstrate any growth modulating effect of IGFBP-3. It is concluded that several agonists acting by nuclear receptors affect IGFBP-3 secretion by LNCaP cells but that the functional significance of these changes warrants further investigation.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Insulin-Like Growth Factor Binding Protein 3/genetics , Metribolone/pharmacology , Prostate/metabolism , Prostatic Neoplasms/genetics , Retinoids/pharmacology , Testosterone Congeners/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Calcitriol/pharmacology , Cell Division/drug effects , Gene Expression Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostate/drug effects , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , Tretinoin/pharmacology , Triiodothyronine/pharmacology , Tumor Cells, Cultured
20.
Mol Cell Endocrinol ; 155(1-2): 69-76, 1999 Sep 10.
Article in English | MEDLINE | ID: mdl-10580840

ABSTRACT

mRNA differential display polymerase chain reaction analysis was used to screen systematically for novel androgen-regulated genes in the human prostatic adenocarcinoma cell line LNCaP. A 232 bp PCR fragment was found to be consistently down-regulated by the synthetic androgen R1881. Sequencing revealed complete identity with the human homologue of mouse Paternally expressed gene 3 (Peg3), an imprinted gene that plays an important role as a downstream mediator of the effects of tumor necrosis factor (TNF). The down-regulation of Peg3 mRNA by androgens was confirmed by Northern blot hybridization. The effect proved time and dose dependent with maximal repression (3.5-fold) after 24 h of treatment with 10(-8) M R1881. The steroid specificity of Peg3 mRNA regulation reflected the aberrant ligand specificity of the mutated androgen receptor in LNCaP cells, supporting the involvement of the androgen receptor in the repression process. Basal expression of Peg3 mRNA was almost completely abolished by the protein synthesis inhibitor cycloheximide. Experiments with Actinomycin D suggested that androgens act at a transcriptional level rather than by changing the stability of Peg3 mRNA. Comparison of the expression of Peg3 mRNA in 50 different human tissues revealed ubiquitous expression, but low levels in the prostate. The highest levels were observed in endocrine tissues such as ovary, placenta, adrenal and pituitary. High levels were also noted in various parts of the brain. No detectable levels of Peg3 mRNA were observed in two other androgen receptor-positive prostate tumor cell lines (MDA PCa-2a and -2b), and in the poorly differentiated and androgen receptor-negative prostate tumor lines PC-3 and DU-145. It is concluded that both androgens and loss of differentiation may affect the expression of Peg3, a mediator of the effects of TNF. Further experiments will be required to explore whether these changes affect the responsiveness of prostate tumor cells to TNF.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Metribolone/pharmacology , Protein Kinases , Proteins/genetics , Testosterone Congeners/pharmacology , Transcription Factors , Transcription, Genetic/drug effects , Adenocarcinoma , Animals , Breast Neoplasms , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Kinetics , Kruppel-Like Transcription Factors , Male , Mice , Polymerase Chain Reaction , Prostatic Neoplasms , RNA, Messenger/genetics , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Tumor Cells, Cultured , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...