Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Oncogene ; 36(10): 1440-1450, 2017 03.
Article in English | MEDLINE | ID: mdl-27694897

ABSTRACT

The presence of intact ligand-binding domain (LBD) ensures the strict androgen-dependent regulation of androgen receptor (AR): binding of androgen induces structural reorganization of LBD resulting in release of AR from HSP90, suppression of nuclear export which otherwise dominates over import and nuclear translocation of AR as a transcription factor. Thus, loss or defects of the LBD abolish constraint from un-liganded LBD as exemplified by constitutively active AR variants (AR-Vs), which are associated with emerging resistance mechanism to anti-AR therapy in castration-resistant prostate cancer (mCRPC). Recent analysis of the AR splicing landscapes revealed mCRPC harboring multiple AR-Vs with diverse patterns of inclusion/exclusion of exons (exons 4-8) corresponding to LBD to produce namely exon-skipping variants. In silico construction for these AR-Vs revealed four novel AR-Vs having unique features: Exclusion of specified exons introduces a frameshift in variants v5es, v6es and v7es. ARv56es maintains the reading frame resulting in the inclusion of the C-terminal half of the LBD. We systematically characterized these AR-Vs regarding their subcellular localization, affinity for HSP90 and transactivation capability. Notably, ARv5es was free from HSP90, exclusively nuclear, and constitutively active similarly as previously reported for v567es. In contrast, v6es and v7es were similar in that they are cytoplasmic, transcriptionally inactive and bind HSP90, ARv56es was present in both nucleus and cytoplasm, does not bind HSP90 and is transcriptionally inactive. Converting these transcriptionally inactive AR-Vs into active forms, we identified the two separate elements that allosterically suppress otherwise constitutively active AR-Vs; one in exon 5 for v6es and v7es and the other in exon 8 for v56es. Our findings identify a novel constitutively active AR-V, ARv5es and establish a method to predict potential activities of AR-Vs carrying impaired LBD.


Subject(s)
Alternative Splicing , Protein Interaction Domains and Motifs/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Cell Line , Exons , Gene Editing , Gene Expression , Genes, Reporter , Genetic Loci , Humans , Intracellular Space , Introns , Ligands , Nonsense Mediated mRNA Decay , Protein Binding , Protein Transport , Receptors, Androgen/chemistry , Transcription, Genetic , Transcriptional Activation
3.
Prostate Cancer Prostatic Dis ; 16(2): 204-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23459096

ABSTRACT

BACKGROUND: Obesity is consistently linked with prostate cancer (PCa) recurrence and mortality, though the mechanism is unknown. Impaired glucose regulation, which is common among obese individuals, has been hypothesized as a potential mechanism for PCa tumor growth. In this study, we explore the relationship between serum glucose at time of treatment and risk of PCa recurrence following initial therapy. METHODS: The study group comprised 1734 men treated with radical prostatectomy (RP) or radiation therapy (RT) for localized PCa between 2001-2010. Serum glucose levels closest to date of diagnosis were determined. PCa recurrence was determined based on PSA progression (nadir PSA+2 for RT; PSA≥0.2 for RP) or secondary therapy. Multivariate Cox regression was performed to determine whether glucose level was associated with biochemical recurrence after adjusting for age, race, body mass index, comorbidity, diagnosis of diabetes, Gleason Sum, PSA, treatment and treatment year. RESULTS: Recurrence was identified in 16% of men over a mean follow-up period of 41 months (range 1-121 months). Those with elevated glucose (≥100 mg/dl) had a 50% increased risk of recurrence (HR 1.5, 95% CI: 1.1-2.0) compared with those with a normal glucose level (<100 mg/dl). This effect was seen in both those undergoing RP (HR 1.9, 95% CI: 1.0-3.6) and those treated with RT (HR 1.4, 95% CI: 1.0-2.0). CONCLUSIONS: Glucose levels at the time of PCa diagnosis are an independent predictor of PCa recurrence for men undergoing treatment for localized disease.


Subject(s)
Hyperglycemia/blood , Neoplasm Recurrence, Local/blood , Prostatic Neoplasms/blood , Aged , Blood Glucose , Brachytherapy , Humans , Hyperglycemia/complications , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/etiology , Neoplasm Recurrence, Local/prevention & control , Obesity/blood , Obesity/complications , Proportional Hazards Models , Prostatectomy , Prostatic Neoplasms/etiology , Prostatic Neoplasms/therapy , Risk
4.
Horm Metab Res ; 44(7): 511-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22495974

ABSTRACT

The insulin-like growth factor-I (IGF-IR) and androgen (AR) receptors are important players in prostate cancer. Functional interactions between the IGF-I and androgen signaling pathways have crucial roles in the progression of prostate cancer from early to advanced stages. DNA methylation is a major epigenetic alteration affecting gene expression. Hypermethylation of tumor suppressor promoters is a frequent event in human cancer, leading to inactivation and repression of specific genes. The aim of the present study was to identify the entire set of methylated genes ("methylome") in a cellular model that replicates prostate cancer progression. The methylation profiles of the P69 (early stage, benign) and M12 (advanced stage, metastatic) prostate cancer cell lines were established by treating cells with the demethylating agent 5-aza-2'-deoxycytidine (5-Aza) followed by DNA microarray analysis. Comparative genome-wide methylation analyses of 5-Aza-treated versus untreated cells identified 297 genes overexpressed in P69 and 191 genes overexpressed in M12 cells. 102 genes were upregulated in both benign and metastatic cell lines. In addition, our analyses identified the PITX2 gene as a master regulator upstream of the AR and IGF-IR genes. The PITX2 promoter was semi-methylated in P69 cells but fully methylated (i. e., silenced) in M12 cells. Epigenetic regulation of PITX2 during the course of the disease may lead to orchestrated control of the AR and IGF signaling pathways. In summary, our results provide new insights into the epigenetic changes associated with progression of prostate cancer from an organ confined, androgen-sensitive disorder to an aggressive, androgen-insensitive disease.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Prostatic Neoplasms/genetics , Receptor, IGF Type 1/genetics , Receptors, Androgen/genetics , Transcription Factors/metabolism , Genes, Neoplasm/genetics , Homeodomain Proteins/genetics , Humans , Male , Middle Aged , Promoter Regions, Genetic/genetics , Receptor, IGF Type 1/metabolism , Receptors, Androgen/metabolism , Transcription Factors/genetics , Homeobox Protein PITX2
5.
Oncogene ; 30(20): 2345-55, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21258401

ABSTRACT

As an established mediator of inflammation, interleukin-6 (IL-6) is implicated to facilitate prostate cancer progression to androgen independence through transactivation of the androgen receptor. However, whether IL-6 has a causative role in de novo prostate tumorigenesis was never investigated. We now provide the first evidence that IL-6 can induce tumorigenic conversion and further progression to an invasive phenotype of non-tumorigenic benign prostate epithelial cells. Moreover, we find that paracrine IL-6 stimulates the autocrine IL-6 loop and autocrine activation of insulin-like type I growth factor receptor (IGF-IR) to confer the tumorigenic property and also that activation of signal transducer and activator of transcription 3 (STAT3) is critical in these processes. Inhibition of STAT3 activation or IGF-IR signaling suppresses IL-6-mediated malignant conversion and the associated invasive phenotype. Inhibition of STAT3 activation suppresses IL-6-induced upregulation of IGF-IR and its ligands, namely IGF-I and IGF-II. These findings indicate that IL-6 signaling cooperates with IGF-IR signaling in the prostate microenvironment to promote prostate tumorigenesis and progression to aggressiveness. Our findings suggest that STAT3 and IGF-IR may represent potential effective targets for prevention or treatment of prostate cancer.


Subject(s)
Interleukin-6/metabolism , Prostatic Neoplasms/metabolism , Autocrine Communication , Cell Line , Cell Transformation, Neoplastic , Disease Progression , Epithelial Cells , Epithelial-Mesenchymal Transition , Humans , Male , Neoplasm Invasiveness , Prostate/cytology , Receptor, IGF Type 1/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcriptional Activation
6.
J Cell Physiol ; 220(2): 376-81, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19360811

ABSTRACT

There is increased interest in the effects of secretory products from aged cells on promoting both benign and malignant cell growth. We identified a human fibroblast line, AG04382, from an aged donor that naturally demonstrated senescence-associated features and whose conditioned media significantly induced proliferation of benign prostatic hyperplasia (BPH1) cells. Candidate cytokines mediating this effect were identified with protein arrays and validated by ELISA. We found that the AG04382 fibroblast line secreted high levels of CXCL5, CCL5, and CCL2, but relative to the other lines, its conditioned media was unique in its increased expression of CCL5. Blocking studies using specific antibodies against CXCL5, CCL5, and CCL2 in the conditioned media of AG04382 showed that only CCL5 contributed significantly to BPH1 proliferation. Stimulation of BPH1 cells with rhuCCL5 resulted in increased proliferation and migration, as well as significant changes in the expression of genes that influence angiogenesis. These data suggest that CCL5 is a candidate chemokine secreted by aged cells that promotes prostate growth and regulates angiogenesis.


Subject(s)
Cell Proliferation , Cellular Senescence/physiology , Chemokine CCL5/metabolism , Epithelial Cells/physiology , Fibroblasts/metabolism , Neovascularization, Physiologic/genetics , Prostate/cytology , Age Factors , Aged, 80 and over , Cell Line , Cell Movement/physiology , Chemokine CCL5/genetics , Culture Media, Conditioned/chemistry , Epithelial Cells/cytology , Fibroblasts/cytology , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Oligonucleotide Array Sequence Analysis , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology
7.
Br J Cancer ; 98(2): 250-5, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18182993

ABSTRACT

Whether tumours are epithelial or non-epithelial in origin, it is generally accepted that once they reach a certain size all solid tumours are dependent upon a vascular supply to provide nutrients. Accordingly, there is great interest in how the extracellular environment enhances or inhibits vascular growth. In this minireview, we will examine key extracellular components, their changes with ageing, and discuss how these alterations may influence the subsequent development of tumour vasculature in the aged host. Because of the tight correlation between advanced age and development of prostate cancer, we will use prostate cancer as the model throughout this review.


Subject(s)
Extracellular Space/physiology , Neoplasms, Glandular and Epithelial/blood supply , Neovascularization, Pathologic/etiology , Prostatic Neoplasms/blood supply , Aged , Aged, 80 and over , Androgens/metabolism , Androgens/physiology , Extracellular Matrix/physiology , Extracellular Matrix Proteins/physiology , Extracellular Space/enzymology , Extracellular Space/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Male , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/physiology , Models, Biological , Neoplasms, Glandular and Epithelial/pathology , Prostatic Neoplasms/pathology
8.
Neurology ; 70(6): 440-8, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-17942819

ABSTRACT

BACKGROUND: Reduced brain insulin signaling and low CSF-to-plasma insulin ratios have been observed in patients with Alzheimer disease (AD). Furthermore, intracerebroventricular or IV insulin administration improve memory, alter evoked potentials, and modulate neurotransmitters, possibly by augmenting low brain levels. After intranasal administration, insulin-like peptides follow extracellular pathways to the brain within 15 minutes. OBJECTIVE: We tested the hypothesis that daily intranasal insulin treatment would facilitate cognition in patients with early AD or its prodrome, amnestic mild cognitive impairment (MCI). The proportion of verbal information retained after a delay period was the planned primary outcome measure. Secondary outcome measures included attention, caregiver rating of functional status, and plasma levels of insulin, glucose, beta-amyloid, and cortisol. METHODS: Twenty-five participants were randomly assigned to receive either placebo (n = 12) or 20 IU BID intranasal insulin treatment (n = 13) using an electronic atomizer, and 24 participants completed the study. Participants, caregivers, and all clinical evaluators were blinded to treatment assignment. Cognitive measures and blood were obtained at baseline and after 21 days of treatment. RESULTS: Fasting plasma glucose and insulin were unchanged with treatment. The insulin-treated group retained more verbal information after a delay compared with the placebo-assigned group (p = 0.0374). Insulin-treated subjects also showed improved attention (p = 0.0108) and functional status (p = 0.0410). Insulin treatment raised fasting plasma concentrations of the short form of the beta-amyloid peptide (A beta 40; p = 0.0471) without affecting the longer isoform (A beta 42), resulting in an increased A beta 40/42 ratio (p = 0.0207). CONCLUSIONS: The results of this pilot study support further investigation of the benefits of intranasal insulin for patients with Alzheimer disease, and suggest that intranasal peptide administration may be a novel approach to the treatment of neurodegenerative disorders.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/drug effects , Cognition Disorders/drug therapy , Insulin/administration & dosage , Plaque, Amyloid/drug effects , Administration, Intranasal , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/blood , Attention/drug effects , Attention/physiology , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Disease Progression , Humans , Neuroprotective Agents/administration & dosage , Neuropsychological Tests , Peptide Fragments/blood , Peptide Fragments/drug effects , Pilot Projects , Plaque, Amyloid/metabolism , Treatment Outcome , Verbal Behavior/drug effects , Verbal Behavior/physiology
9.
Neurology ; 66(10): 1506-10, 2006 May 23.
Article in English | MEDLINE | ID: mdl-16717209

ABSTRACT

BACKGROUND: Hyperinsulinemia and insulin resistance are risk factors for memory impairment and Alzheimer disease (AD). Insulin regulates levels of the amyloid beta-peptide (Abeta) in vitro in neuronal cultures and in vivo in the CSF of normal older adults. OBJECTIVE: To determine whether insulin affected plasma Abeta levels and whether such effects differed for patients with AD compared with normal older adults. METHODS: Fifty-nine patients with AD and 50 healthy older adults each received infusions of saline and of insulin (1.0 mU.kg(-1).min(-1)) with accompanying dextrose to maintain euglycemia. A subset of participants (19 AD, 12 normal) received two additional conditions, in which insulin was infused at a lower (0.33 mU.kg(-1).min(-1)) and higher (1.67 mU.kg(-1).min(-1)) rate. Plasma insulin and Abeta were measured after 120 minutes of infusion. RESULTS: Adults with AD had higher plasma insulin vs normal adults at the two higher infusion rates, despite receiving comparable amounts of insulin. For normal adults, insulin reduced plasma Abeta levels at the middle (1.0 mU.kg(-1).min(-1)) dose, with attenuated effects at lower and higher doses. In contrast, for patients with AD, insulin raised plasma Abeta levels at the two higher doses (1.0 and 1.67 mU.kg(-1).min(-1)). CONCLUSIONS: These results suggest that patients with Alzheimer disease (AD) have reduced insulin clearance and insulin-provoked plasma amyloid beta-peptide (Abeta) elevation. Abnormal regulation of peripheral Abeta by insulin may contribute to AD risk.


Subject(s)
Alzheimer Disease/blood , Amyloid beta-Peptides/blood , Insulin/pharmacology , Peptide Fragments/blood , Aged , Aged, 80 and over , Apolipoproteins E/genetics , Body Mass Index , Dose-Response Relationship, Drug , Female , Genotype , Glucose Clamp Technique , Humans , Hyperinsulinism/blood , Insulin/administration & dosage , Male , Middle Aged
10.
Neurobiol Aging ; 27(3): 451-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-15964100

ABSTRACT

Raising insulin acutely in the periphery and in brain improves verbal memory. Intranasal insulin administration, which raises insulin acutely in the CNS without raising plasma insulin levels, provides an opportunity to determine whether these effects are mediated by central insulin or peripheral processes. Based on prior research with intravenous insulin, we predicted that the treatment response would differ between subjects with (epsilon4+) and without (epsilon4-) the APOE-epsilon4 allele. On separate mornings, 26 memory-impaired subjects (13 with early Alzheimer's disease and 13 with amnestic mild cognitive impairment) and 35 normal controls each underwent three intranasal treatment conditions consisting of saline (placebo) or insulin (20 or 40 IU). Cognition was tested 15 min post-treatment, and blood was acquired at baseline and 45 min after treatment. Intranasal insulin treatment did not change plasma insulin or glucose levels. Insulin treatment facilitated recall on two measures of verbal memory in memory-impaired epsilon4- adults. These effects were stronger for memory-impaired epsilon4- subjects than for memory-impaired epsilon4+ subjects and normal adults. Unexpectedly, memory-impaired epsilon4+ subjects showed poorer recall following insulin administration on one test of memory. These findings suggest that intranasal insulin administration may have therapeutic benefit without the risk of peripheral hypoglycemia and provide further evidence for apolipoprotein E (APOE) related differences in insulin metabolism.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Apolipoproteins E/genetics , Cognition/drug effects , Insulin/administration & dosage , Memory Disorders/drug therapy , Memory Disorders/genetics , Aged , Alzheimer Disease/epidemiology , Comorbidity , Female , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Humans , Incidence , Male , Memory Disorders/epidemiology , Risk Assessment/methods , Risk Factors , Treatment Outcome , Washington/epidemiology
11.
Leukemia ; 19(4): 580-5, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15703779

ABSTRACT

The pathophysiology of the myelodysplastic syndromes (MDS) is incompletely understood. Tumor necrosis factor (TNF)alpha levels are elevated, particularly in early-stage MDS, and apoptosis in marrow cells is upregulated. Observations in other models have shown a role for insulin-like growth factor binding protein 3 (IGFBP-3) in TNFalpha-mediated apoptosis. We observed increased levels of IGFBP-3 in the marrow plasma of patients with MDS (P = 0.005) and hypothesized that altered IGFBP-3 levels contribute to the dysregulation of hemopoiesis in MDS by affecting proliferation and apoptosis. Western analysis of marrow plasma from MDS patients revealed an increase in the ratio of intact vs fragmented IGFBP-3 in early-stage MDS (relative to controls) that decreased with MDS disease progression, suggesting increased proteolysis with more advanced disease. Thus, these results provide evidence for dysregulation of IGFBP-3 in patients with MDS. While the data are complex, they are consistent with a modulatory effect of IGFBP-3 on hemopoiesis in MDS. Conceivably, understanding these mechanisms may allow for the development of novel therapeutic strategies.


Subject(s)
Apoptosis/physiology , Bone Marrow/metabolism , Hematopoiesis/physiology , Insulin-Like Growth Factor Binding Protein 3/metabolism , Myelodysplastic Syndromes/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Hematopoiesis/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Membrane Glycoproteins/metabolism , Middle Aged , Myelodysplastic Syndromes/pathology , Myelodysplastic Syndromes/physiopathology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/metabolism
12.
Horm Metab Res ; 35(11-12): 751-7, 2003.
Article in English | MEDLINE | ID: mdl-14710354

ABSTRACT

The type I insulin-like growth factor receptor (IGF-IR) plays a critical role in signaling survival and proliferation in many cell types. Activation of IGF-IR by its ligands promotes cell proliferation via mitogen-activated protein kinase (MAPK) cascade and cell survival via phosphoinositide 3-kinase (PI3K) cascade. The IGF-IR emerges as a powerful growth factor for many tumor cells. A truncated IGF-IR 486/STOP, described as a dominant negative IGF-IR mutant, was shown to induce apoptosis and inhibit tumor growth in vivo while endogenous IGF-IR was activated. To investigate the mechanism(s) of the action of 486/STOP, we have introduced 486/STOP into the prostate tumor model cell line M12 and its derivative M12lisn that expresses high levels of wild type IGF-IR. We have found that 486/STOP induces apoptosis in M12 and M12lisn cells in culture and that 486/STOP acts through activation of the pro-apoptotic p38-MAPK without interfering with wild type IGF-IR activation. In addition, our results have indicated that 486/STOP induced activation of p38-MAPK increases through activation of endogenous IGF-IR. These data suggest that activation of the IGF-IR by 486/STOP can selectively enhance the previously reported IGF-IR pro-apoptotic signaling pathways.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Receptor, IGF Type 1/physiology , Apoptosis , Cell Division , Cell Line, Tumor , Cell Survival , Enzyme Activation , Humans , Male , Receptor, IGF Type 1/genetics , Sequence Deletion , p38 Mitogen-Activated Protein Kinases
13.
J Endocrinol ; 170(3): 609-18, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11524241

ABSTRACT

The biologically active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) has been shown to regulate the proliferation of human prostate epithelial cell lines. Since the insulin-like growth factor (IGF) system is involved in the transformation process of epithelial cells, the following study was undertaken to determine if the IGF system, in particular IGF binding protein-3 (IGFBP-3), is altered by 1,25-(OH)2D3 in normal prostate epithelial cells as part of a mechanism for inhibition of transformation. Two cell systems were used in this study: (1) primary cultures of benign human prostate epithelial cells (PECs) and (2) an SV40-T immortalized prostate epithelial cell line (P153) that is non-tumorigenic. 1,25-(OH)2D3 was added to parallel sets of PECs and P153 cells in addition to the presence or absence of IGF-I or des(1-3)IGF-I. Treatment with 1,25-(OH)2D3 resulted in significant growth inhibition of both PECs and P153 cells. Furthermore, 1,25-(OH)2D3 inhibited IGF-induced proliferation, but this was partially reversed by high concentrations of IGF-I. Western ligand blots of condition media demonstrated a significant increase in IGFBP-3; likewise Northern blots demonstrated an increase in mRNA for IGFBP-3. Proliferation assays using an antibody designed to block the IGF-independent effects of IGFBP-3 failed to reverse the inhibitory effect of 1,25-(OH)2D3. Thus, IGFBP-3 acts in an IGF-dependent manner to inhibit cell growth of benign prostate epithelial cells.


Subject(s)
Calcitriol/pharmacology , Epithelial Cells/drug effects , Insulin-Like Growth Factor Binding Protein 3/metabolism , Prostate/drug effects , Cell Culture Techniques , Cell Division/drug effects , Cell Line , Cell Transformation, Viral , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/immunology , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/pharmacology , Male , Peptide Fragments/pharmacology , Prostate/cytology , Prostate/metabolism
14.
Neurology ; 57(4): 605-12, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11524467

ABSTRACT

OBJECTIVE: To characterize the cognitive and neuroendocrine response to treatment with a high dose of estrogen for postmenopausal women with AD. METHODS: Twenty postmenopausal women with AD were randomized to receive either 0.10 mg/day of 17 beta-estradiol by skin patch or a placebo patch for 8 weeks. Subjects were evaluated at baseline, at weeks 3, 5, and 8 during treatment, and again 8 weeks after treatment termination. During each visit, cognition was assessed with a battery of neuropsychological tests, and blood samples were collected to measure plasma estradiol as well as several other neuroendocrine markers of interest. RESULTS: Significant effects of estrogen treatment were observed on attention (Stroop Color Word Interference Test), verbal memory (Buschke Selective Reminding Test), and visual memory (Figure Copy/Memory). In addition, women treated with estrogen demonstrated improved performance on a test of semantic memory (Boston Naming Test) compared with subjects who received a placebo. Estrogen appeared to have a suppressive effect on the insulin-like growth factor (IGF) system such that plasma concentration of IGF binding protein-3 was significantly reduced and plasma levels of estradiol and IGF-I were negatively correlated during estrogen treatment. CONCLUSIONS: Administration of a higher dose of estrogen may enhance attention and memory for postmenopausal women with AD. Although these findings provide further clinical evidence to support a cognitive benefit of estrogen for women with AD, studies evaluating the effect of estradiol administration, in particular, using larger sample sizes and for longer treatment durations are warranted before the therapeutic potential of estrogen replacement for women with AD can be firmly established.


Subject(s)
Alzheimer Disease/drug therapy , Cognition/drug effects , Estradiol/administration & dosage , Estrogen Replacement Therapy/methods , Insulin-Like Growth Factor I/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/psychology , Analysis of Variance , Cognition/physiology , Double-Blind Method , Estradiol/blood , Female , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor II/metabolism , Middle Aged
15.
Endocrinology ; 142(5): 1990-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11316765

ABSTRACT

Insulin-like growth factor-binding proteins (IGFBPs) both stimulate and inhibit IGF activity, and in the M12 prostate cancer cell line, overexpression of IGFBP-4 was shown to delay tumorigenesis while decreasing the production of IGFBP-2. We have performed the reverse experiment, inhibition of IGFBP-4 expression with antisense complementary DNA, in two prostate tumor cell lines, ALVA-31 and M12. Expression of antisense messenger RNA transcripts was verified by RNase protection assays, and inhibition of mature IGFBP-4 in cell medium was demonstrated by Western blotting. Both transfected lines (ALVA-31asBP4 and M12asBP4) proliferated more slowly in monolayer culture than parental controls. Colony formation in soft agar was strongly inhibited in both cases, and the rate of tumor formation and growth in male athymic nude mice injected with M12asBP4 was markedly reduced relative to that in mice receiving M12 control cells. Apoptosis induced by the topoisomerase inhibitor etoposide was also enhanced in transfected cells. The effects on colony formation in soft agar and tumor formation in mice were maintained for the duration of the experiments, in contrast to the delayed growth observed in the previous study of IGFBP-4 overexpression. A significant difference was found in the patterns of IGFBP expression; production of both messenger RNA and protein for IGFBP-3 and IGFBP-6 was greatly increased in the M12asBP4 and ALVA31asBP4 cell lines. Up-regulation of these binding proteins has been observed in association with actions of 1,25-dihydroxyvitamin D(3) in prostate cancer cells, and the data suggest a role for IGFBP-3 and IGFBP-6 in the suppression of prostate tumor cell growth.


Subject(s)
DNA, Antisense/therapeutic use , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor Binding Protein 4/physiology , Insulin-Like Growth Factor Binding Protein 6/biosynthesis , Prostatic Neoplasms/therapy , Animals , Apoptosis , Cell Division , Humans , Insulin-Like Growth Factor Binding Protein 4/antagonists & inhibitors , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transfection , Tumor Cells, Cultured
16.
Endocrinology ; 142(1): 21-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145562

ABSTRACT

A marked decrease in the type 1 insulin-like growth factor (IGF) receptor (IGF-IR) occurs in prostate epithelial cells during transformation from the benign to the metastatic state. One of the principal regulators of IGF-IR gene expression, the WT1 tumor suppressor, is expressed in prostate cancer and in prostate cancer cell lines. The purpose of this study was to determine whether the decrease in IGF-IR expression was transcriptionally regulated, and whether WT1 action may be involved in the repression of the IGF-IR gene in prostate cancer cells. The P69 cell line was derived by immortalization of human primary prostate epithelial cells with simian virus-40 T antigen and is rarely tumorigenic. The M12 line was derived from the P69 line by selection for tumor formation in nude mice and is tumorigeneic and metastatic. P69 cells express 20,000 IGF-IR/cell, whereas M12 cells express 3,500 IGF-IR/cell. These differences in receptor number are reflected in proportional differences in IGF-IR mRNA levels. To assess IGF-IR promoter activity in these cell lines, each was transiently transfected with luciferase reporter vectors containing the IGF-IR gene transcription start site and 476 bp of 5'-flanking sequence, 640 bp of 5'-untranslated region sequence, or both regions. The promoter activity of the full-length construct was 50% lower (P < 0.01) in M12 cells compared with P69 cells, the activity of the 5'-flanking region construct was 53% lower (P < 0.0001), and that of the 5'-untranslated region construct was 36% lower (P = 0.01). P69 clones stably transfected with a WT1 expression vector exhibited decreased expression of the endogenous IGF-IR gene and decreased promoter activity in transient transfection assays with IGF-IR promoter constructs containing multiple WT1 binding sites. The observed reduction in endogenous IGF-IR expression was sufficient to inhibit IGF-I-stimulated cell proliferation. These data suggest that most of the decreased expression of the IGF-IR seen in malignant prostate epithelium is the result of transcriptional repression of the IGF-IR gene, and that this repression may be due in part to the increased expression of the WT1 tumor suppressor in metastatic prostate cancer.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor I/pharmacology , Prostatic Neoplasms/genetics , Receptor, IGF Type 1/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Division/drug effects , Cell Line, Transformed , DNA-Binding Proteins/analysis , Genes, Wilms Tumor , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/pathology , Recombinant Fusion Proteins/analysis , Simian virus 40/genetics , Transcription Factors/analysis , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured , WT1 Proteins
17.
Endocrinology ; 141(11): 4072-80, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11089538

ABSTRACT

Insulin-like growth factor (IGF)-binding protein (IGFBP)-related proteins (IGFBP-rPs) are newly described cysteine-rich proteins that share significant aminoterminal structural similarity with the conventional IGFBPs and are involved in a diversity of biological functions, including growth regulation. IGFBP-rP1 (MAC25/Angiomodulin/prostacyclin-stimulating factor) is a potential tumor-suppressor gene that is differentially expressed in meningiomas, mammary and prostatic cancers, compared with their malignant counterparts. We have previously shown that IGFBP-rP1 is preferentially produced by primary cultures of human prostate epithelial cells (HPECs) and by poorly tumorigenic P69SV40T cells, compared with the cancerous prostatic LNCaP, DU145, PC-3, and M12 cells. We now show that IGFBP-rP1 increases during senescence of HPEC. IGFBP-rP2 (also known as connective tissue growth factor), a downstream effector of transforming growth factor (TGF)-beta and modulator of growth for both fibroblasts and endothelial cells, was detected in most of the normal and malignant prostatic epithelial cells tested, with a marked up-regulation of IGFBP-rP2 during senescence of HPEC. Moreover, IGFBP-rP2 noticeably increased in response to TGF-beta1 and all-trans retinoic acid (atRA) in HPEC and PC-3 cells, and it decreased in response to IGF-I in HPEC. IGFBP-rP3 [nephroblastoma overexpressed (NOV)], the protein product of the NOV protooncogene, was not detected in HPEC but was expressed in the tumorigenic DU145 and PC-3 cells. It was also synthesized by the SV40-T antigen-transformed P69 and malignant M12 cells, where it was down-regulated by atRA. These observations suggest biological roles of IGFBP-rPs in the human prostate. IGFBP-rP1 and IGFBP-rP2 are likely to negatively regulate growth, because they seem to increase during senescence of the prostate epithelium and in response to growth inhibitors (TGF-beta1 and atRA). Although the data collected on IGFBP-rP3 in prostate are modest, its role as a growth stimulator and/or protooncogene is supported by its preferential expression in cancerous cells and its down-regulation by atRA.


Subject(s)
Cellular Senescence , Insulin-Like Growth Factor Binding Protein 1/physiology , Insulin-Like Growth Factor Binding Protein 2/physiology , Insulin-Like Growth Factor Binding Protein 3/physiology , Prostate/cytology , Animals , Cell Division , Cell Line , Epithelial Cells/chemistry , Epithelial Cells/cytology , Gene Expression , Humans , Insulin-Like Growth Factor Binding Protein 1/analysis , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 2/analysis , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 3/analysis , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/pharmacology , Male , Mice , Prostate/chemistry , RNA, Messenger/analysis , Time Factors , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Tumor Cells, Cultured
19.
Electrophoresis ; 21(9): 1823-31, 2000 May.
Article in English | MEDLINE | ID: mdl-10870968

ABSTRACT

Several methods have been developed for the comprehensive analysis of gene expression in complex biological systems. Generally these procedures assess either a portion of the cellular transcriptome or a portion of the cellular proteome. Each approach has distinct conceptual and methodological advantages and disadvantages. We have investigated the application of both methods to characterize the gene expression pathway mediated by androgens and the androgen receptor in prostate cancer cells. This pathway is of critical importance for the development and progression of prostate cancer. Of clinical importance, modulation of androgens remains the mainstay of treatment for patients with advanced disease. To facilitate global gene expression studies we have first sought to define the prostate transcriptome by assembling and annotating prostate-derived expressed sequence tags (ESTs). A total of 55000 prostate ESTs were assembled into a set of 15953 clusters putatively representing 15953 distinct transcripts. These clusters were used to construct cDNA microarrays suitable for examining the androgen-response pathway at the level of transcription. The expression of 20 genes was found to be induced by androgens. This cohort included known androgen-regulated genes such as prostate-specific antigen (PSA) and several novel complementary DNAs (cDNAs). Protein expression profiles of androgen-stimulated prostate cancer cells were generated by two-dimensional electrophoresis (2-DE). Mass spectrometric analysis of androgen-regulated proteins in these cells identified the metastasis-suppressor gene NDKA/nm23, a finding that may explain a marked reduction in metastatic potential when these cells express a functional androgen receptor pathway.


Subject(s)
Databases, Factual , Gene Expression , Prostate/metabolism , Proteome/genetics , DNA, Complementary , Expressed Sequence Tags , Gene Expression Profiling/methods , Humans , Male , Mass Spectrometry , Oligonucleotide Array Sequence Analysis
20.
Psychoneuroendocrinology ; 24(6): 657-77, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10399774

ABSTRACT

Preliminary evidence from clinical studies indicates that treatment with estrogen augments cognitive function for women with Alzheimer's disease (AD). The neurobiology of estrogen, particularly its neuromodulatory and neuroprotective actions, provide a viable basis to support such cognition-enhancing effects. We conducted a placebo-controlled, double-blind, parallel-group design pilot clinical study to evaluate the cognitive and neuroendocrine response to estrogen administration for postmenopausal women with AD. Twelve women with probably AD of mild-moderate severity completed the study. During an eight week treatment period, six women received 0.05 mg/day dosage of 17 beta-estradiol via a skin patch and the remaining six wore a placebo skin patch. Subjects were randomized to equal distribution, and evaluated at baseline, at weeks 1, 3, 5, and 8 on treatment, and at weeks 9, 10, 11, and 13 off treatment. On each day of evaluation, cognition was assessed using a battery of neuropsychological tests, and blood samples were collected to measure plasma concentrations of estradiol and estrone. In addition, several neuroendocrine markers were measured in plasma to evaluate the relationship between estrogen-induced cognitive effects and fluctuations in the catecholaminergic and insulin-like growth factor systems. Significant effects of estrogen treatment were observed on attention (i.e. Stroop: number of self-corrections in the Interference condition, F[1,8] = 8.22, P < 0.03) and verbal memory (i.e., Buschke: delayed cued recall, F[3,30] = 4.31, P < 0.02). The salutary effects of estrogen on cognition were observed after the first week of treatment, and started to diminish when treatment was terminated. For women treated with estrogen, enhancement in verbal memory was positively correlated with plasma levels of estradiol (r = 0.96, P < 0.02) and negatively correlated with concentrations of insulin-like growth factor binding protein-3 (IGFBP-3) in plasma (r = -0.92, P < 0.03). Furthermore, a trend in the data was evident to suggest a negative relationship between plasma levels of insulin-like growth factor-1 (IGF-1) and verbal memory (r = -0.86, P = 0.06). Estrogen administration suppressed peripheral markers of the IGF system, as evidenced by a negative correlation between plasma concentration of estradiol and IGF-1 (r = -0.93, P < 0.03), and a trend for a similar relationship between plasma levels of estradiol and IGFBP-3 (r = -0.86, P = 0.06). With respect to the catecholamines assayed, norepinephrine was positively correlated with verbal memory (r = 0.95, P < 0.02) for women who were treated with estrogen. Furthermore, there was a trend to suggest a negative relationship between plasma epinephrine levels and the number of errors committed on a test of attention (r = -0.84, P = 0.07). In the placebo group, no significant effects of estrogen replacement were evident either on measures of cognition or on any of the neuroendocrine markers. The results of this study suggest that estrogen replacement may enhance cognition for postmenopausal women with AD. Furthermore, several markers of neuroendocrine activity may serve to index the magnitude of estrogen-induced facilitation on cognition. In addition, research findings from the present study will provide important information for the design of larger prospective clinical studies that are essential to definitively establish the therapeutic role of estrogen replacement for postmenopausal women with AD.


Subject(s)
Alzheimer Disease/drug therapy , Climacteric/drug effects , Estrogen Replacement Therapy , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/metabolism , Neuropsychological Tests , Norepinephrine/blood , Administration, Cutaneous , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/psychology , Double-Blind Method , Estradiol/administration & dosage , Estradiol/blood , Estrone/blood , Female , Humans , Mental Recall/drug effects , Pilot Projects , Verbal Learning/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...