Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
JACC Basic Transl Sci ; 9(3): 364-379, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38559622

ABSTRACT

Low testosterone (T), common in aging men, associates with cardiovascular disease. We investigated whether follicle-stimulating hormone (FSH), which is affected by T, modulates the cardiovascular effects associated with low T or castration. FSHß-/-:low-density lipoprotein receptor (LDLR)-/- mice, untreated or castrated (orchiectomy, gonadotropin-releasing hormone agonist or antagonist), demonstrated significantly less atherogenesis compared with similarly treated LDLR-/- mice, but not following FSH delivery. Smaller plaque burden in LDLR-/- mice receiving gonadotropin-releasing hormone antagonists vs agonists were nullified in FSHß-/-:LDLR-/- mice. Atherosclerotic and necrotic plaque size and macrophage infiltration correlated with serum FSH/T. In patients with prostate cancer, FSH/T following androgen-deprivation therapy initiation predicted cardiovascular events. FSH facilitates cardiovascular disease when T is low or eliminated.

3.
J Urol ; 207(5): 1020-1028, 2022 05.
Article in English | MEDLINE | ID: mdl-34978211

ABSTRACT

PURPOSE: Cardiovascular disease is a common cause of death in prostate cancer patients. Low testosterone is associated with increased cardiovascular risk in the general male population. We investigated the relationship between serum testosterone, cardiovascular disease and risk factors in androgen-deprivation therapy-naïve prostate cancer patients. MATERIALS AND METHODS: We performed a cross-sectional analysis of a subgroup of 1,326 androgen-deprivation therapy-naïve men from RADICAL-PC (Role of Androgen-Deprivation Therapy In CArdiovascular Disease-A Longitudinal Prostate Cancer study) in whom serum testosterone was measured at baseline. RADICAL-PC is a prospective multicenter cohort study of men (2,565) enrolled within 1 year of prostate cancer diagnosis, or within 6 months of commencing androgen-deprivation therapy for the first time. Cardiovascular risk factors, cancer characteristics and total serum testosterone were collected at baseline. Low testosterone was defined as total serum testosterone <11 nmol/L (<320 ng/dL). A Framingham cardiovascular risk score ≥15 was considered high risk for future cardiovascular events. We performed logistic regression to calculate odds ratios for the association between testosterone and cardiovascular risk. RESULTS: Among 1,326 participants (median age 67 years, range 45-93), 553 (42%) had low testosterone. Low testosterone was associated with existing cardiovascular disease, diabetes, elevated hemoglobin A1c, obesity, hypertriglyceridemia, low high-density lipoprotein cholesterol, hypertension and Framingham score >15. Among patients with low testosterone, the odds ratio for high cardiovascular risk was 1.33 (1.02-1.73) after adjusting for ethnicity, education, alcohol use, cancer characteristics, physical activity and body mass index. CONCLUSIONS: Among androgen-deprivation therapy-naïve prostate cancer patients, low testosterone is common and associated with increased cardiovascular risk factors.


Subject(s)
Cardiovascular Diseases , Prostatic Neoplasms , Aged , Aged, 80 and over , Androgen Antagonists/adverse effects , Androgens , Cardiovascular Diseases/complications , Cardiovascular Diseases/etiology , Cohort Studies , Cross-Sectional Studies , Humans , Male , Middle Aged , Prospective Studies , Testosterone
5.
Prostate Cancer Prostatic Dis ; 24(2): 389-397, 2021 06.
Article in English | MEDLINE | ID: mdl-32989260

ABSTRACT

BACKGROUND: Unlike in other mouse models of atherogenesis, it has recently been suggested that orchiectomy plays a role in accelerating atherosclerosis and inhibiting the progression of cardiovascular disease in the ApoE-/-:Ins2+/Akita mouse model of hyperglycemia. Androgen-deprivation therapy (ADT) is a common treatment for prostate cancer, a population with high prevalence of cardiovascular disease and its risk factors. Our objectives were to test and further characterize the effects of pharmacological castration which is currently the acceptable modality to deliver ADT in the clinic. METHODS: Male ApoE-/-:Ins2+/Akita mice received one of three modes of ADT (gonadotropin-releasing hormone (GnRH)-antagonist (degarelix), GnRH-agonist (leuprolide), or bilateral orchiectomy) and were compared to corresponding untreated control mice (n = 9-13/group). Mice were followed for 5 months. Body weight, fasting blood glucose, glucose tolerance, serum C-peptide, leptin, and testosterone levels along with atherosclerotic aortic plaque size and characteristics were determined. In a separate experiment, the survival of mice, untreated and on ADT, was determined. RESULTS: Castration was achieved for all three modes of ADT. However, degarelix-treated mice gained significantly less weight, had lower serum leptin levels and systolic blood pressure compared to orchiectomy and leuprolide-treated mice. ADT improved dysglycemia and atherosclerotic burden. GnRH-antagonist significantly improved survival compared to GnRH-agonist but not compared to orchiectomy. CONCLUSIONS: Further characterization of the ApoE-/-:Ins2+/Akita mouse model confirms that pharmacological ADT ameliorated metabolic syndrome and cardiovascular complications. Improved dysglycemia and atherosclerosis associated with increased survival which was longest after degarelix followed by orchiectomy.


Subject(s)
Androgen Antagonists/pharmacology , Cardiovascular Diseases/prevention & control , Disease Models, Animal , Hyperglycemia/complications , Metabolic Syndrome/prevention & control , Orchiectomy/methods , Protective Factors , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Insulin/physiology , Leuprolide/pharmacology , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/pathology , Mice , Mice, Knockout, ApoE/physiology , Oligopeptides/pharmacology
7.
J Urol ; 202(6): 1199-1208, 2019 12.
Article in English | MEDLINE | ID: mdl-31188734

ABSTRACT

PURPOSE: Androgen deprivation therapy may increase the risk of cardiovascular disease. Limited data suggest that GnRH (gonadotropin-releasing hormone) antagonist may be associated with a lower risk of cardiovascular disease than GnRH agonist. MATERIALS AND METHODS: We performed a phase II, randomized, open label study in men with prostate cancer and preexisting cardiovascular disease who were randomized to receive GnRH agonists or antagonists for 1 year. The primary outcome was endothelial function measured by the EndoPAT 2000 device (Itamar Medical, Caesarea, Israel). The predefined secondary outcome was a new cardiovascular event. Patients were followed for the development of cardiovascular disease, defined as death, myocardial infarction, a cerebrovascular event, percutaneous angioplasty with coronary stent insertion or hospitalizations due to cardiac events. RESULTS: A total of 80 patients were enrolled in study, including 41 and 39 who received GnRH antagonist and agonist, respectively. Patients in each arm had similar baseline characteristics. We did not detect a difference in the primary end point (endothelial function) between the groups (mean ± SD reactive hyperemia index 2.07 ± 0.15 vs 1.92 ± 0.11, p=0.42). However, during the trial period a new cardiovascular event (the secondary end point) developed in 15 patients. Of cases new major cardiovascular and cerebrovascular events developed in 9, including death in 2, myocardial infarction in 1, a cerebrovascular event in 2 and percutaneous angioplasty with coronary stent insertion in 4. Of the patients 20% randomized to GnRH agonist experienced a major cardiovascular and cerebrovascular event compared to 3% of those on GnRH antagonist (p=0.013). The absolute risk reduction in major cardiovascular and cerebrovascular events at 12 months using GnRH antagonist was 18.1% (95% CI 4.6-31.2, p=0.032). CONCLUSIONS: To our knowledge this is the first prospective study to test cardiovascular outcomes among patients with prostate cancer who received androgen deprivation therapy. No differences in the primary end point were noted between the study arms. However, the secondary end point revealed that patients treated with GnRH agonist experienced significantly more major cardiovascular and cerebrovascular events than those treated with GnRH antagonist. These phase II results suggest that in patients with prostate cancer who have preexisting cardiovascular disease selecting the androgen deprivation therapy modality may differentially affect cardiac outcomes.


Subject(s)
Androgen Antagonists/adverse effects , Cardiovascular Diseases/epidemiology , Prostatic Neoplasms/drug therapy , Aged , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/complications , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Humans , Incidence , Male , Pilot Projects , Prospective Studies , Prostatic Neoplasms/complications
8.
Bladder Cancer ; 4(2): 139-147, 2018 Apr 26.
Article in English | MEDLINE | ID: mdl-29732385

ABSTRACT

Patients with bladder cancer are at high risk of developing both venous and arterial thromboembolic events. Factors that contribute to this phenomenon include the hypercoagulable state induced by the malignancy itself, medical comorbidities that are common in this predominantly elderly patient population as well as treatments such as prolonged pelvic surgery and cisplatin-based chemotherapy. While formal guidelines address prevention of venous thromboembolism in patients undergoing radical cystectomy, consensus regarding the role of pharmacologic prophylaxis in patients with bladder cancer being treated with chemotherapy, either with neoadjuvant or adjuvant intent in conjunction with radical cystectomy, as part of bladder preservation protocols or for metastatic disease, has proved elusive. The present narrative review was undertaken to define the incidence of and identify risk factors for thromboembolism among patients with bladder cancer, as well as to assess the efficacy of pharmacologic prophylaxis in reducing the risk of thromboembolism in this patient population.

9.
Oncol Lett ; 13(5): 3624-3630, 2017 May.
Article in English | MEDLINE | ID: mdl-28521463

ABSTRACT

Endoplasmic reticulum (ER) protein ERp46 is a member of the protein disulfide isomerase family of oxidoreductases, which facilitates the reduction of disulfides in proteins and their folding. Accumulation of misfolded proteins has been implicated in cancer. The objectives of the present study were to investigate the role of ERp46 in prostate cancer, its expression and its effects on prostate cancer growth. A tissue microarray with human prostate cancer and normal prostate tissue samples was stained for ERp46 followed by image analysis. Human prostate adenocarcinoma 22Rv1 cells were stably transfected with short hairpin RNA (shRNA) specific for ERp46, a non-effective scrambled control or a plasmid containing full-length human ERp46 cDNA, and cell growth was determined. Subcloned cells were treated with thapsigargin or tunicamycin to induce ER stress and lysates were subjected to western blot analysis for ER stress proteins. Subcutaneous xenografts of parental 22Rv1, ERp46-overexpressing (ERp46+), shERp46 or scrambled control cells were established in male inbred BALB/c nude mice (n=10/group). Tumor growth curves of the xenografts were constructed over a period of 30 days and subsequently the mice were sacrificed and the amount of serum prostate-specific antigen was determined. The results demonstrated increased ERp46 expression levels in prostate cancer tissue samples of Gleason ≥7 compared with normal prostate tissue samples. When ERp46 was stably knocked down using shRNA or overexpressed in prostate carcinoma 22Rv1 cells, tumor growth in vitro and in BALB/c nude mice was inhibited and accelerated, respectively. ERp46 overexpression led to reduced sensitivity to ER stress as indicated by higher half maximal inhibitory concentrations for tunicamycin and thapsigargin in ERp46+ cells. The shERp46 cells lost the ability to upregulate protein disulfide isomerase following tunicamycin-induced ER stress. The present study suggests a role for ERp46 as a therapeutic target in prostate cancer, given its expression profile in human prostate cancer, and its effect on prostate cancer cell growth.

11.
Prostate ; 77(2): 211-221, 2017 02.
Article in English | MEDLINE | ID: mdl-27699825

ABSTRACT

BACKGROUND: Prostate cancer development is associated with numerous lifestyle factors (i.e., physical activity, nutrition intake) and metabolic perturbations. These factors have been studied independently; here, we used an integrative approach to characterize these lifestyle and metabolic parameters in men undergoing diagnostic prostate biopsies. METHODS: We prospectively evaluated 51 consecutive men for body composition, metabolic factors including glucose- and lipid-related measures, as well as lifestyle factors prior to prostate biopsy. Evaluations were performed in a blinded manner and were subsequently related to biopsy outcomes for: (i) presence or absence of cancer; and (ii) where cancer was present, Gleason score. RESULTS: Serum C-peptide concentrations were significantly greater in participants with Gleason scores ≥4 + 3 (2.8 ± 1.1 ng/ml) compared to those with Gleason 3 + 3 (1.4 ± 0.6 ng/ml) or Gleason 3 + 4 (1.3 ± 0.8 ng/ml, P = 0.002), suggesting greater insulin secretion despite lack of differences in fasting glucose concentrations. Central adiposity, measured by waist circumference, was significantly greater in participants with Gleason ≥4 + 3 (110.1 ± 7.4 cm) compared to those with Gleason 3 + 4 (102.0 ± 9.5 cm, P = 0.028). Men with Gleason ≥4 + 3 also had significantly greater leptin concentrations than those with lower Gleason scores (Gleason ≥4 + 3: 15.6 ± 3.3 ng/ml vs. Gleason 3 + 4: 8.1 ± 8.1 ng/ml, P < 0.05) and leptin:adiponectin ratio (Gleason ≥4 + 3: 9.7 ± 6.1 AU, Gleason 3 + 4: 2.9 ± 3.2, Gleason 3 + 3: 2.4 ± 2.1 AU, P = 0.013). CONCLUSIONS: We profiled a cluster of obesity-related metabolic perturbations (C-peptide, central adiposity, leptin, and leptin:adiponectin ratios) which may associate with more aggressive prostate cancer histology. Prostate 77:211-221, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adipokines/blood , Biomarkers, Tumor/blood , C-Peptide/blood , Obesity, Abdominal/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/diagnosis , Aged , Aged, 80 and over , Body Composition/physiology , Humans , Male , Middle Aged , Neoplasm Grading , Obesity, Abdominal/diagnosis , Prospective Studies , Risk Factors , Waist Circumference/physiology
12.
J Urol ; 196(6): 1627-1633, 2016 12.
Article in English | MEDLINE | ID: mdl-27312316

ABSTRACT

PURPOSE: Neoadjuvant chemotherapy and pelvic surgery are significant risk factors for thromboembolic events. Our study objectives were to investigate the timing, incidence and characteristics of thromboembolic events during and after neoadjuvant chemotherapy and subsequent radical cystectomy in patients with muscle invasive bladder cancer. MATERIALS AND METHODS: We performed a multi-institutional retrospective analysis of 761 patients who underwent neoadjuvant chemotherapy and radical cystectomy for muscle invasive bladder cancer from 2002 to 2014. Median followup from diagnosis was 21.4 months (range 3 to 272). Patient characteristics included the Khorana score, and the incidence and timing of thromboembolic events (before vs after radical cystectomy). Survival was calculated using the Kaplan-Meier method. The log rank test and multivariable Cox proportional hazards regression were used to compare survival between patients with vs without thromboembolic events. RESULTS: The Khorana score indicated an intermediate thromboembolic event risk in 88% of patients. The overall incidence of thromboembolic events in patients undergoing neoadjuvant chemotherapy was 14% with a wide variation of 5% to 32% among institutions. Patients with thromboembolic events were older (67.6 vs 64.6 years, p = 0.02) and received a longer neoadjuvant chemotherapy course (10.9 vs 9.7 weeks, p = 0.01) compared to patients without a thromboembolic event. Of the thromboembolic events 58% developed preoperatively and 72% were symptomatic. On multivariable regression analysis the development of a thromboembolic event was not significantly associated with decreased overall survival. However, pathological stage and a high Khorana score were adverse risk factors for overall survival. CONCLUSIONS: Thromboembolic events are common in patients with muscle invasive bladder cancer who undergo neoadjuvant chemotherapy before and after radical cystectomy. Our results suggest that a prospective trial of thromboembolic event prophylaxis during neoadjuvant chemotherapy is warranted.


Subject(s)
Chemotherapy, Adjuvant/adverse effects , Cystectomy/adverse effects , Thromboembolism/epidemiology , Urinary Bladder Neoplasms/therapy , Aged , Chemotherapy, Adjuvant/methods , Cystectomy/methods , Female , Humans , Incidence , Male , Middle Aged , Neoplasm Invasiveness , Retrospective Studies , Survival Analysis , Thromboembolism/etiology , Urinary Bladder/pathology , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
13.
Oncol Rep ; 34(4): 1968-76, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26252502

ABSTRACT

While normal kidneys are relatively sensitive to ionizing radiation (IR), renal cell carcinoma (RCC) is considered radioresistant. Carbonic anhydrase IX (CA9), an enzyme that maintains intracellular pH by carbon dioxide dissolution, is upregulated in the majority of RCC, but not in normal kidneys. Since regulation of intracellular pH may enhance radiation effects, we hypothesized that inhibition of CA9 may radiosensitize RCC. Clonogenic survival assay of human clear cell RCC 786-O and murine RCC RAG cells in the presence of a pharmacological CA9 inhibitor or with shRNA-mediated knockdown of CA9 was performed to investigate the response to IR in vitro (single dose or fractionated) and in vivo. Extracellular pH changes were measured in vitro. Treatment with AEBS [4-(2-aminoethyl)benzene sulfonamide], a sulfonamide, was used as a pharmacological inhibitor of the enzymatic activity of CA9. Nude mice bearing subcutaneous xenografts of 786-O cells stably expressing CA9 shRNA or scrambled control were irradiated (6 Gy). Tumor growth was followed longitudinally in the 786-O-bearing mice receiving AEBS (50-200 µg/ml drinking water) or control (vehicle only) which were irradiated (6 Gy) and compared with mice receiving either IR or AEBS alone. In vitro inhibition of CA9 activity or expression significantly sensitized RCC cells to the effects of IR (p<0.05), an effect even more significant when hypofractionated IR was applied. In vivo irradiated xenografts from RCC cells transfected with CA9 shRNA were significantly smaller compared to irradiated xenografts from the scrambled shRNA controls (p<0.05). RCC xenografts from mice treated with AEBS in combination with IR grew significantly slower than all controls (p<0.05). Inhibition of CA9 expression or activity resulted in radiation sensitization of RCC in a preclinical mouse model.


Subject(s)
Antigens, Neoplasm/genetics , Carbonic Anhydrases/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/radiotherapy , Radiation Tolerance/genetics , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/drug effects , Carbonic Anhydrase IX , Carbonic Anhydrases/biosynthesis , Carbonic Anhydrases/drug effects , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , RNA, Small Interfering/genetics , Radiation, Ionizing , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays
14.
Urol Oncol ; 32(8): 1126-34, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25242517

ABSTRACT

OBJECTIVES: Observational studies relate androgen deprivation therapy (ADT) to metabolic syndrome (MS) and cardiovascular disease, an association potentially subject to uncontrollable confounding factors, especially diet and genetic/metabolic risk factors. In the absence of prospective randomized clinical trials, causality remains unproven. We comparatively investigated the effects of different ADT modalities on the development of MS and atherosclerosis in a mouse model. MATERIALS AND METHODS: Low-density lipoprotein receptor knockout mice underwent orchiectomy plus vehicle (2.5% mannitol), sham surgery plus vehicle (control), sham surgery plus gonadotropin-releasing hormone (GnRH) antagonist (degarelix), or sham surgery plus GnRH agonist (leuprolide) (n = 9-13/group) and were followed for 4 months. Visceral fat accumulation, lean body mass, adipocyte size, fasting blood glucose, glucose tolerance, serum levels of leptin, follicle-stimulating hormone, luteinizing hormone, and testosterone, along with atherosclerotic plaque size and characteristics were measured. RESULTS: All 3 modes of ADT decreased circulating testosterone levels in mice, although leuprolide treatment reached nadir levels of testosterone later. Orchiectomized and leuprolide-treated mice gained significantly more visceral fat compared with degarelix-treated mice. Improved glucose tolerance tests were recorded in degarelix-treated mice. The aortic atherosclerotic plaque area in leuprolide-treated and orchiectomized mice was larger than in control mice (P<0.005 and P = 0.002, respectively), but it was not significantly different from control in degarelix-treated mice. The necrotic core area in degarelix-treated mice was smaller compared with leuprolide-treated and orchiectomized mice (P = 0.011 and P = 0.002, respectively). CONCLUSIONS: Our results suggest that ADT induced MS and atherosclerosis in a preclinical mouse model to a mode-specific extent. GnRH antagonist generated the least atherosclerosis and characteristics of MS compared with orchiectomy and GnRH agonist.


Subject(s)
Androgen Antagonists/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Atherosclerosis/etiology , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Metabolic Syndrome/etiology , Adipose Tissue/pathology , Adiposity , Androgen Antagonists/adverse effects , Animals , Antineoplastic Agents, Hormonal/adverse effects , Atherosclerosis/chemically induced , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Disease Models, Animal , Humans , Leuprolide/administration & dosage , Leuprolide/adverse effects , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/pathology , Metabolic Syndrome/prevention & control , Mice , Mice, Knockout , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Orchiectomy/adverse effects , Orchiectomy/methods , Random Allocation , Receptors, LDL/genetics , Receptors, LDL/metabolism
15.
PLoS One ; 9(3): e90389, 2014.
Article in English | MEDLINE | ID: mdl-24594673

ABSTRACT

An established inverse clinical correlation between serum adiponectin levels and renal cell carcinoma (RCC) aggressiveness exists. We have recently demonstrated that adiponectin suppresses clear cell RCC (ccRCC) progression through interaction with its receptor, adiponectin receptor 1 (AdipoR1). ERp46 has been shown to inhibit adiponectin signaling via interaction with AdipoR1 in HeLa cells. However, the expression of ERp46 in RCC has not been described thus far. The objectives of this study were to investigate ERp46 in RCC, its expression, its effects on RCC growth in a mouse model and whether it interacts with AdipoR1. We demonstrated a higher ERp46/AdipoR1 expression ratio in metastatic compared to non-metastatic ccRCC, as determined by immunohistochemistry of tissue microarrays and subsequent image analysis. When ERp46 was stably knocked down using shRNA or overexpressed in murine RCC RAG cells, RCC growth after subcutaneous injection in BALB/c nude mice was inhibited and accelerated, respectively. In vitro analysis to determine the molecular interaction between AdipoR1 and ERp46 included co-immunoprecipitation using human ccRCC 786-O cells and a bacterial adenylate cyclase-based two hybrid system and demonstrated no sustained AdipoR1-ERp46 interaction. This is the first report to suggest a role for ERp46 as a potential therapeutic target in RCC given its expression profile in human RCC samples and its effect on in vivo RCC growth. Since a stable interaction with AdipoR1 could not be established, we suggest that the tumorigenic properties of ERp46 in RCC cells are not related to an inhibitory modulation of AdipoR1.


Subject(s)
Carcinoma, Renal Cell/metabolism , Endoplasmic Reticulum/metabolism , Kidney Neoplasms/metabolism , Protein Disulfide-Isomerases/metabolism , Animals , Blotting, Western , Carcinoma, Renal Cell/pathology , Humans , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude
16.
Clin Exp Metastasis ; 31(2): 169-83, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24096711

ABSTRACT

Energy-sensing pathways, normally coordinated by 5' AMP-activated protein kinase (AMPK), are dysregulated in renal cell carcinoma (RCC). Obesity can accentuate the pre-existing pro-tumorigenic metabolic machinery in RCC cells through its associated obesogenic hormonal milieu, characterized by lower circulating levels of adiponectin. In RCC patients, low adiponectin levels associate clinically with more aggressive disease. We investigated the adiponectin signaling pathway in RCC, focusing on adiponectin receptor 1 (AdipoR1) and associated activation of AMPK. AdipoR1 protein in RCC and normal surrounding renal tissues was determined by Western blot analysis and immunohistochemistry. Anti-tumorigenic effects of adiponectin in RCC cells in vitro were investigated via VEGF and MMP ELISA and invasion assays. Using in vivo models of RCC, the effect of AdipoR1-knockdown (shRNA) on tumor latency, growth and dissemination were determined. AdipoR1 protein was significantly reduced in clear cell RCC specimens. Adiponectin treatment inhibited VEGF, MMP-2 and MMP-9 secretion and activity and invasive and migratory capacities of RCC cells. AMPKα1-knockdown (shRNA) attenuated adiponectin's effects. In cells stably expressing AdipoR1-specific shRNA, AMPK activation by adiponectin was significantly reduced compared to cells expressing control shRNA. In vivo, AdipoR1 knockdown increased the growth, dissemination and angiogenesis of RCC. These findings suggest that deficiencies in the entire adiponectin hormonal axis (the hormone and its receptor) result in underactivation of AMPK leading to increased angiogenic and invasive capacities of RCC. The established link between obesity and RCC can therefore be further explained by the adiponectin deficiency in obese individuals together with reduced AdipoR1 protein in RCC.


Subject(s)
Adiponectin/physiology , Carcinoma, Renal Cell/physiopathology , Kidney Neoplasms/physiopathology , Receptors, Adiponectin/physiology , AMP-Activated Protein Kinase Kinases , Adiponectin/genetics , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Humans , Kidney Neoplasms/pathology , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinases/metabolism , Receptors, Adiponectin/genetics , Vascular Endothelial Growth Factor A/metabolism
17.
Anticancer Drugs ; 20(2): 115-22, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19209028

ABSTRACT

Commonly used chemotherapeutic agents for breast cancer treatment include cisplatin, doxorubicin, and paclitaxel. Unfortunately, these effective antiproliferative agents are limited by their toxicities. Previously, we have shown that doxycycline can substantially reduce tumor burden in an animal model of breast cancer bone metastasis. The purpose of this study was to examine the effect of doxycycline in combination with chemotherapy. Human breast adenocarcinoma MDA-MB-231 cells were treated in vitro with each drug individually and in combination with doxycycline. Cell survival was determined using the clonogenic survival assay. Doxycycline in combination with doxorubicin or paclitaxel yielded therapeutic antagonism at all effect levels. Combinatory treatment with cisplatin, however, yielded a biphasic interaction, at low combinatorial doses, the effect was quantified as nearly additive, whereas higher doxycycline-cisplatin doses yielding greater than 50% cell inhibition resulted in synergistic effects. Cell cycle profiles were determined and showed that treatment with doxycycline, cisplatin, and doxorubicin resulted in G1-phase, S-phase, and G2/M-phase arrests, respectively. Upon addition of doxycycline to doxorubicin, the G2/M-arrest characteristic of doxorubicin-only treatment was abrogated, which may account for the observed antagonism. Cells treated with doxycycline and cisplatin showed a further increase in S-phase arrest, also observed with cisplatin alone, which may be responsible for the additive and synergistic effects on cell survival. We clearly show that doxycycline in combination with paclitaxel or doxorubicin treatment resulted in antagonism; however, combining doxycycline with cisplatin led to synergistic interactions at higher effect levels. The increased potency of cisplatin may warrant dose reduction and thus decrease toxicity in vivo.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Doxycycline/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Interactions , Drug Synergism , Female , Humans , Inhibitory Concentration 50 , Paclitaxel/pharmacology
18.
Oncol Rep ; 20(4): 897-903, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18813833

ABSTRACT

Patients with advanced prostate cancer frequently have a poor prognosis as a result of metastasis. The serum prostate-specific antigen (PSA) test is widely used for the diagnosis of prostate cancer. The enzymatic activity of PSA may be involved in the invasion of prostate cancer. We set out to determine the prevalent form of PSA in human prostate adenocarcinoma samples by ELISA and Western blot analysis and its enzymatic activity using a synthetic substrate S-2586 and fibronectin. Our results show that in serum from prostate cancer patients and in tumour homogenates, the prevalent form was PSA bound to alpha1-antichymotrypsin. All homogenates showed enzymatic activity towards a synthetic PSA substrate, whereas only five samples showed activity at 28 kDa towards fibronectin as determined by enzymography, which is most likely due to active PSA. Human prostate cancer LNCaP cells produced largely inactive PSA. In comparison, 22Rv1 cells produced 29-fold less PSA, but with high specific activity. Similarly, our results from the human prostate cancer tissue samples also show that free PSA appears to exist in diverse forms of very different specific activity. Since PSA, as a serine protease, may be involved in the invasion of prostate cancer, our results suggest that prostate cancers have potentially diverse invasive capacity due to differences in specific enzymatic activity of PSA.


Subject(s)
Fibronectins/metabolism , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/chemistry , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Male , Middle Aged , Prostate-Specific Antigen/physiology
19.
J Trace Elem Med Biol ; 22(3): 215-23, 2008.
Article in English | MEDLINE | ID: mdl-18755397

ABSTRACT

Although dietary silicon (Si) is recognized to be an important factor for the growth and development of bone and connective tissue, its biochemical role has yet to be identified. The predominant Si-containing species in blood and other biofluids is orthosilicic acid, Si(OH)(4). Dimethylsilanediol, (CH(3))(2)Si(OH)(2), is an environmental contaminant that results from decomposition of silicone compounds used in personal hygiene, health care and industrial products. We examined the in vitro effects of both Si species on the survival (colony forming efficiency), proliferation (DNA content), differentiation (alkaline phosphatase activity) and adhesion (relative protein content) of the human osteoblast-like cell lines Saos-2 and hFOB 1.19. Orthosilicic acid yielded a small, dose-dependent decrease in Saos-2 cell survivability up to its 1,700 micromol/L solubility limit, by which point survival was 20% less than that of untreated cells. This negative association, although small, correlated with a reduction in the proliferation and adhesion of Saos-2 cells as well as of hFOB 1.19 and osteoclast-like GCT cells. By contrast, dimethylsilanediol treatment had no discernable influence on Saos-2 survivability at concentrations up to 50 micromol/L, and yet significantly enhanced cell survival at higher doses. Moreover, dimethylsilanediol did not affect proliferation or adhesion of any cell line. The findings show that orthosilicic acid and dimethylsilanediol affect osteoblast-like cells very differently, providing insight into the mechanism by which silicon influences bone health, although the specific site of Si activity remains unknown. There was no evidence to suggest that dimethylsilanediol is cytotoxic at environmental/physiological concentrations.


Subject(s)
Organosilicon Compounds/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Silicic Acid/pharmacology , Alkaline Phosphatase/metabolism , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Integrin alphaVbeta3/metabolism , Osteoblasts/metabolism
20.
Clin Exp Metastasis ; 23(5-6): 249-58, 2006.
Article in English | MEDLINE | ID: mdl-17086360

ABSTRACT

Numerous cancer patients fail standard chemotherapy or develop resistance to chemotherapy during the course of treatment. The purpose of this study is to elucidate the overall response of cells obtained from cancer patients and from normal individuals to chemotherapeutic agents. We analysed the chemosensitivity of cancer cells derived from bone marrow and from pleural effusions or ascites fluids from patients with different cancers. Chemosensitivity to doxorubicin, cisplatin and paclitaxel was determined using the MTT assay. We also determined the response of bone marrow mononuclear (BMMN) cells. There was a wide range of responses to chemotherapy drugs in samples from different individuals. This was observed in cells derived from bone marrow and from ascites or pleural fluids. Large variations were also observed among morphologically normal BMMN cells and metastatic cancer cells from chemo-naïve patients. Cancer cells can easily be collected from ascites or pleural fluids and reliably assayed for chemosensitivity. We describe here that inherent chemoresistance may be a reason for the lack of response to chemotherapy in some patients. We discuss the potential of using the determination of natural resistance to dictate the drugs to be employed for treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Marrow Cells/drug effects , Drug Resistance, Neoplasm , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/drug effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Ascitic Fluid/cytology , Bone Marrow Cells/pathology , Cell Survival , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Female , Formazans/analysis , Humans , Inhibitory Concentration 50 , Male , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Pleural Effusion, Malignant/cytology , Tetrazolium Salts/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...