Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
J Intellect Disabil Res ; 67(4): 352-361, 2023 04.
Article in English | MEDLINE | ID: mdl-36543755

ABSTRACT

BACKGROUND: Historically, individuals with Down syndrome have been excluded from clinical research. Our objective was to assess the degree of interest adults with Down syndrome have in participating in research from the perspective of the caregivers who care for them. METHODS: We conducted an online survey of N = 390 caregivers of adults with Down syndrome and asked about interest in research participation and demographics. RESULTS: Caregivers were mostly family members, older than 55 years, and White. Caregivers reported that the adult with Down syndrome that they cared for would be more comfortable participating in research that was physiological, such as research involving fit bits (70.2% would participate), exercise (63.3%) or diet apps (53.9%), whereas they would be less likely to participate in clinical trials involving more invasive procedures such as injections (10.9%) and laboratory exams like MRIs (32.0%). We found little difference by age or gender of the adult with Down syndrome or by caregiver education level. CONCLUSIONS: Our survey identified high interest for less invasive studies, illustrating acceptability of observational and lifestyle studies. More effort may be needed to understand fear and barriers to participation and to create tools and methods to increase interest in more invasive studies.


Subject(s)
Caregivers , Down Syndrome , Humans , Adult , Family , Surveys and Questionnaires , Perception
4.
Cell Commun Signal ; 19(1): 78, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34284799

ABSTRACT

The urinary tract is highly innervated by autonomic nerves which are essential in urinary tract development, the production of growth factors, and the control of homeostasis. These neural signals may become dysregulated in several genitourinary (GU) disease states, both benign and malignant. Accordingly, the autonomic nervous system is a therapeutic target for several genitourinary pathologies including cancer, voiding dysfunction, and obstructing nephrolithiasis. Adrenergic receptors (adrenoceptors) are G-Protein coupled-receptors that are distributed throughout the body. The major function of α1-adrenoceptors is signaling smooth muscle contractions through GPCR and intracellular calcium influx. Pharmacologic intervention of α-and ß-adrenoceptors is routinely and successfully implemented in the treatment of benign urologic illnesses, through the use of α-adrenoceptor antagonists. Furthermore, cell-based evidence recently established the antitumor effect of α1-adrenoceptor antagonists in prostate, bladder and renal tumors by reducing neovascularity and impairing growth within the tumor microenvironment via regulation of the phenotypic epithelial-mesenchymal transition (EMT). There has been a significant focus on repurposing the routinely used, Food and Drug Administration-approved α1-adrenoceptor antagonists to inhibit GU tumor growth and angiogenesis in patients with advanced prostate, bladder, and renal cancer. In this review we discuss the current evidence on (a) the signaling events of the autonomic nervous system mediated by its cognate α- and ß-adrenoceptors in regulating the phenotypic landscape (EMT) of genitourinary organs; and (b) the therapeutic significance of targeting this signaling pathway in benign and malignant urologic disease. Video abstract.


Subject(s)
Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, beta-1/genetics , Urologic Diseases/genetics , Urologic Neoplasms/genetics , Adrenergic beta-Antagonists/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Humans , Male , Prostate/metabolism , Prostate/pathology , Signal Transduction/drug effects , Tumor Microenvironment/genetics , Urinary Tract/metabolism , Urinary Tract/pathology , Urologic Diseases/pathology , Urologic Neoplasms/pathology
7.
Exp Oncol ; 34(3): 165-75, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23070001

ABSTRACT

Emergence of castration-resistant metastatic prostate cancer is due to activation of survival pathways, including apoptosis suppression and anoikis resistance, and increased neovascularization. Thus targeting of apoptotic players is of critical significance in prostate cancer therapy since loss of apoptosis and resistance to anoikis are critical in aberrant malignant growth, metastasis and conferring therapeutic failure. The majority of therapeutic agents act through intrinsic mitochondrial, extrinsic death receptor pathways or endoplasmic reticulum stress pathways to induce apoptosis. Current therapeutic strategies target restoring regulatory molecules that govern the pro-survival pathways such as PTEN which regulates AKT activity. Other strategies focus on reactivating the apoptotic pathways either by down-regulating anti-apoptotic players such as BCL-2 or by up-regulating pro-apoptotic protein families, most notably, the caspases. Caspases are a family of cystine proteases which serve critical roles in apoptotic and inflammatory signaling pathways. During tumorigenesis, significant loss or inactivation of lead members in the caspase family leads to impairing apoptosis induction, causing a dramatic imbalance in the growth dynamics, ultimately resulting in aberrant growth of human cancers. Recent exploitation of apoptosis pathways towards re-instating apoptosis induction via caspase re-activation has provided new molecular platforms for the development of therapeutic strategies effective against advanced prostate cancer as well as other solid tumors. This review will discuss the current cellular landscape featuring the caspase family in tumor cells and their activation via pharmacologic intervention towards optimized anti-cancer therapeutic modalities. This article is part of a Special Issue entitled "Apoptosis: Four Decades Later".


Subject(s)
Apoptosis/genetics , Caspases/metabolism , Prostatic Neoplasms , Signal Transduction , Animals , Humans , Male , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
8.
J Inherit Metab Dis ; 32(2): 289-96, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19277893

ABSTRACT

Phenylketonuria (PKU) is an autosomal recessive disorder resulting in neurological and intellectual disability when untreated. However, even in treated patients there may be residual neurological impairment such as tremor. It has been suggested that the hyperphenylalaninaemia in patients with PKU reduces complex I (NADH:ubiquinone reductase) activity of the mitochondrial respiratory chain (MRC) and/or biosynthesis of coenzyme Q(10) (CoQ(10)), which acts as an electron carrier in the MRC, leading to impaired energy metabolism in the brain of patients with PKU and hence the neurological pathology. The aim of this study was to elucidate the mechanism of phenylalanine (Phe) toxicity on the MRC. We compared mean plasma and blood-spot Phe and mononuclear CoQ(10) levels in 17 patients with PKU and a tremor compared to 22 patients without tremor. Human 1321N1 astrocytoma cells were exposed to hyperphenylalaninaemia by the addition of 300 or 900 micromol/L of Phe to the cell culture medium. Following 96 h of culture we measured complex I and citrate synthase activities and CoQ(10) level. Results showed no significant difference in Phe or CoQ(10) levels in patients with tremor compared to those without tremor. Further, hyperphenylalaninaemia did not cause a significant reduction in complex I activity or CoQ(10) biosynthesis, even when taking into account the mitochondrial enrichment of the cell samples by expressing complex I and CoQ(10) as a ratio to citrate synthase. In conclusion, the results of this study suggest that hyperphenylalaninaemia does not contribute to the pathophysiology of PKU by causing a decrease in MRC complex I activity and/or CoQ(10) biosynthesis.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Electron Transport/physiology , Mitochondrial Diseases/metabolism , Phenylalanine/blood , Adult , Amino Acid Metabolism, Inborn Errors/blood , Cell Line, Tumor , Cells, Cultured , Culture Media , Female , Humans , Lactic Acid/metabolism , Male , Middle Aged , Phenylketonurias/blood , Phenylketonurias/metabolism , Pyruvic Acid/metabolism , Tremor/blood , Tremor/etiology , Tyrosine/blood , Ubiquinone/analogs & derivatives , Ubiquinone/blood , Young Adult
9.
Oncogene ; 27(57): 7171-9, 2008 Dec 04.
Article in English | MEDLINE | ID: mdl-18931702

ABSTRACT

Hypoxia has been previously linked to the development of both benign prostatic hyperplasia and prostate cancer. This study investigated the effect of maspin, an extracellular matrix (ECM) tumor suppressor, on the apoptotic response of prostate cancer cells to hypoxia. Gene expression profiling of human benign and malignant prostate epithelial cells after exposure to hypoxia or normoxia revealed dramatic changes in ECM regulators. Maspin was found to be overexpressed in response to hypoxia in prostate cancer cells, but not in benign prostate cells. To dissect the contribution of maspin to tumor cell responses within a hypoxic microenvironment, we used maspin-overexpressing DU-145 human prostate cancer cells. Exposure to hypoxic conditions (1% O(2)) led to a significant increase in apoptosis in the DU-145 maspin cells, compared to DU-145 neo-transfectants without a significant effect on cell migration. This enhanced sensitivity to hypoxia-induced apoptosis leads to a significant suppression of tumor growth and tumor vascularity in vivo by targeting Akt and focal adhesion kinase activation. Our findings implicate maspin in prostate cancer cell response to hypoxia via recruitment of intracellular signaling partners. This study may have significance in the identification of maspin-driven therapeutic targeting in advanced metastatic prostate cancer.


Subject(s)
Apoptosis/physiology , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serpins/metabolism , Animals , Blotting, Western , Cell Adhesion/physiology , Cell Hypoxia , Cell Movement/physiology , Cell Proliferation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Signal Transduction/physiology
10.
Inflamm Res ; 54(9): 380-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16273337

ABSTRACT

OBJECTIVE: To examine whether HIV-1 Tat protein increases the metastatic potential of human breast cancer cells through induction of pro-inflammatory tumor microenvironment. METHODS: Real-time RT-PCR and ELISA were employed to determine the mRNA and protein expression of IL-6 and IL-8 in highly metastatic human breast cancer cell line, MDA-MB-231. To investigate the transcriptional regulatory mechanisms of Tat-mediated up-regulation of IL-6 and IL-8, EMSA and reporter gene assay were carried out. RESULTS: Exposure of MDA-MB-231 cells to Tat resulted in a significant and dose-dependent up-regulation of IL-6 and IL-8 mRNA and protein expression. HIV-1 Tat protein also markedly increased NF-kappaB DNA-binding activity and transactivation in MDA-MB-231 cells. Additionally, pretreatment with NF-kappaB inhibitors significantly attenuated the ability of Tat to up-regulate IL-6 and IL-8 expression. It was also found that exposure of MDA-MB-231 cells to Tat induced up-regulation of MMP-9 expression at both mRNA and protein levels. CONCLUSIONS: These results suggest that HIV-1 Tat protein up-regulates expression of IL-6 and IL-8 in human breast cancer cells by NF-kappaB-dependent pathway. These data may contribute to exploration of the new molecular mechanisms leading to novel approaches for the therapeutic drug developments specifically targeted against the inflammatory pathways of breast cancer metastasis in AIDS patients.


Subject(s)
Breast Neoplasms/metabolism , Gene Products, tat/pharmacology , Interleukin-6/metabolism , Interleukin-8/metabolism , Cell Line, Tumor , DNA/metabolism , Humans , Matrix Metalloproteinase 9/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Transcriptional Activation/drug effects , Up-Regulation , tat Gene Products, Human Immunodeficiency Virus
11.
Article in English | MEDLINE | ID: mdl-15477876

ABSTRACT

Early diagnosis of prostate cancer holds tremendous promise for the effective therapy and impact on survival of prostate cancer patients. High-grade prostatic intraepithelial neoplasia (HGPIN) is generally accepted as a lesion indicative of a late pathological event in the premalignant changes leading to full development of prostate cancer. This review seeks to identify specific molecular events that may be linked directly to the molecular transition from benign prostate epithelial cells to prostate carcinoma. HGPIN is pathologically detected in a limited group of men undergoing prostate cancer screening for an elevated serum prostate-specific antigen (PSA) or abnormal digital rectal examination (DRE). Loss of apoptotic control provides a molecular basis for the contribution of specific defective steps in the pathway towards development and progression of prostate cancer. Comparative dissection of the apoptosis status and expression profile of key apoptotic regulators among foci of highly proliferative benign prostatic epithelium, PIN and prostate adenocarcinoma from adjacent areas of the same gland revealed a novel insight into the dysfunctional apoptosis events contributing to prostate carcinogenesis. The sequential and notable loss of the three critical signaling components of the apoptotic action of transforming growth factor-beta (TGF-beta), in the prostate, that is, the transmembrane receptor II (TbetaRII), the key cell cycle inhibitor p27(Kip1), as well as the protagonist downstream effector of the TGF-beta signaling mechanism, Smad4, points to their potential value to 'faithfully' characterize HGPIN, as a premalignant prostate lesion. Recent evidence on the molecular changes in apoptosis regulators contributing to HGPIN and their role as molecular markers of disease onset, as well as candidates for therapeutic targeting/chemoprevention of prostate cancer in its early stages will be discussed.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Apoptosis , Biomarkers/analysis , Prostatic Intraepithelial Neoplasia/diagnosis , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Adenocarcinoma/prevention & control , Cell Proliferation , Cell Transformation, Neoplastic , Diagnosis, Differential , Growth Substances , Humans , Male , Physical Examination , Prognosis , Prostate-Specific Antigen/analysis , Prostatic Intraepithelial Neoplasia/prevention & control , Prostatic Neoplasms/prevention & control , Rectum , Signal Transduction
12.
Br J Cancer ; 88(10): 1615-21, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12771931

ABSTRACT

Previous studies documented the ability of quinazoline-based alpha1-adrenoceptor antagonists to induce apoptosis in prostate cancer cells via an alpha 1-adrenoceptor-independent mechanism. In this study we investigated the molecular events initiating this apoptotic effect. Since transforming growth factor-beta 1 (TGF-beta 1) mediates prostate epithelial cell apoptosis, we hypothesised that the activation of the TGF-beta 1 pathway underlies the quinazoline-based apoptotic effect in prostate cancer cells. Treatment of the androgen-independent human prostate cancer cells PC-3 with doxazosin resulted in a strong caspase-3 activation within 24 h, whereas tamsulosin, a sulphonamide-based alpha 1-adrenoceptor antagonist, had no significant apoptotic effect against prostate cancer cells. To identify the molecular components involved in this quinazoline-mediated apoptosis, cDNA microarray analysis of PC-3 prostate cancer cells treated with doxazosin (3 h) was performed. Induced expression of several genes was observed including p21(WAF-1) and I kappa B alpha (inhibitor of NF-kappa B alpha). Relative quantitative reverse transcription-polymerase chain reaction analysis revealed induction of several TGF-beta1 signalling effectors: Induction of mRNA for Smad4 and the TGF-beta1-regulated apoptosis-inducing transcription factor TGF-beta1-inducible early gene (TIEG1) was detected within the first 6 h of doxazosin treatment. Upregulation of I kappa B alpha at both the mRNA and protein level was also detected after 6 h of treatment. Furthermore, doxazosin resulted in a considerable elevation in Smad4 and TIEG protein expression (6 h). A 'latent' increase in TGF-beta mRNA expression was detected after 48 h of treatment. These findings suggest that the quinazoline-based doxazosin mediates prostate cancer apoptosis by initially inducing the expression of TGF-beta1 signalling effectors and subsequently I kappa B alpha. The present study provides an initial insight into the molecular targets of the apoptotic action of quinazolines against prostate cancer cells.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Apoptosis/drug effects , Doxazosin/pharmacology , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , Quinazolines/pharmacology , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/physiology , Transforming Growth Factor beta/pharmacology , AMP-Activated Protein Kinase Kinases , Caspase 3 , Caspases/pharmacology , Humans , Male , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/pharmacology , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
13.
Prostate Cancer Prostatic Dis ; 5(2): 88-95, 2002.
Article in English | MEDLINE | ID: mdl-12496995

ABSTRACT

alpha(1)-Adrenoceptor antagonists, have been documented to induce apoptosis and reduce prostate tumor vascularity in benign and malignant prostate cells. The quinazoline based alpha(1)-antagonists, doxazosin and terazosin but not tamsulosin (a sulphonamide derivative) suppress prostate growth without affecting cell proliferation. These quinazoline-mediated apoptotic effects occur via an alpha(1)-adrenoceptor independent mechanism potentially involving activation of the TGF-beta signal transduction pathway. This review discusses the current knowledge of the action of quinazoline-derived alpha(1)-adrenoceptor antagonists in the benign and malignant prostate and their potential therapeutic use in the treatment of benign prostatic hyperplasia (BPH) and prostate cancer. Finally, a molecular pathway is proposed for their observed apoptotic function against prostate cells. Increased understanding of the action of these established and clinically accepted agents would provide a basis for the design of safe, effective therapeutic regimens in the treatment of prostatic diseases.


Subject(s)
Adrenergic Antagonists/pharmacology , Apoptosis , Prostatic Hyperplasia/physiopathology , Prostatic Neoplasms/physiopathology , Quinazolines/pharmacology , Receptors, Adrenergic, alpha-1/physiology , Humans , Male , Signal Transduction , Tumor Cells, Cultured
14.
Article in English | MEDLINE | ID: mdl-11562954

ABSTRACT

The ring-expanded ("fat") nucleoside, 4,8-diamino-6-imino-6H-1-beta-D-ribofuranosylimidazo[4,5-e][1,3]diazepine (1) and its 2',3',5'-tri-O-benzoyl derivative (2) exhibited potent broad spectrum anticancer activities in vitro against a wide variety of human tumor cell lines. The tribenzoyl derivative 2 was found to be considerably more active than the parent nucleoside 1. Further studies using human prostate cancer cells PC-3 and DU-145 suggest that the treatment of exponentially growing culture cells with 1 and 2 leads to marked loss of cell viability in a dose-dependent manner.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Purine Nucleosides/pharmacology , Antineoplastic Agents/chemical synthesis , Azepines/chemical synthesis , Drug Screening Assays, Antitumor , Humans , Male , Nucleosides/pharmacology , Prostatic Neoplasms/drug therapy , Purine Nucleosides/chemical synthesis , Tumor Cells, Cultured
15.
Prostate ; 48(2): 71-8, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11433417

ABSTRACT

BACKGROUND: We previously demonstrated that the quinazoline-derived a1-adrenoceptor antagonists doxazosin and terazosin suppress prostate cancer growth via apoptosis induction. The aim of this study was to determine the potential effect of a1-adrenoceptor antagonists on tumor vascularity of the human prostate. METHODS: A total of 34 men with benign prostatic hyperplasia (BPH) who have been on terazosin treatment (for the obstructive symptoms) were pathologically diagnosed with prostate cancer following surgery. These patients were stratified according to the length of treatment periods with terazosin into two groups, 1 week-6 months, and 6-17 months. The control group consisted of prostatectomy specimens from 25 untreated prostate cancer patients undergoing surgery for localized disease. Formalin-fixed, paraffin-embedded prostate specimens were analyzed for apoptosis (TUNEL assay), cell proliferation (Ki-67), microvessel density (MVD) (von Willebrand factor/Factor VIII), vascular endothelial growth factor (VEGF) expression, and prostate specific antigen (PSA) immunoreactivity. RESULTS: A significant induction of apoptosis was observed among cancerous prostatic epithelial cells in the terazosin-treated, as compared to the untreated prostate cancer specimens, while there was no significant change in the proliferative index of the same tumor cell populations after treatment. Furthermore, terazosin resulted in a significant decrease in prostate tissue MVD compared with the untreated group (P < 0.01), that correlated with the increased apoptotic index of the cancerous areas. Tissue PSA expression in the prostatic tumor foci was also markedly reduced after terazosin treatment, while no significant changes in VEGF expression were detected. CONCLUSIONS: These findings provide the first evidence that terazosin, a quinazoline-based a1-blocker decreases prostate tumor vascularity. Our study has significant clinical implications in identifying selected alpha1-adrenoceptor antagonists as potential anti-tumor agents with apoptotic and anti-angiogenic effects in the human prostate that can be exploited for the treatment of advanced prostate cancer.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Neovascularization, Pathologic , Prazosin/pharmacology , Prostatic Neoplasms/physiopathology , Aged , Aged, 80 and over , Apoptosis , Cell Division , Endothelial Growth Factors/analysis , Endothelial Growth Factors/biosynthesis , Factor VIII/analysis , Humans , Lymphokines/analysis , Lymphokines/biosynthesis , Male , Middle Aged , Prazosin/analogs & derivatives , Prostate-Specific Antigen/biosynthesis , Prostatic Hyperplasia , Retrospective Studies , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Endocrinology ; 142(6): 2419-26, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356690

ABSTRACT

In this study, the potential interactions between dihydrotestosterone (DHT), a survival factor, and transforming growth factor-beta (TGF-beta), an apoptotic inducer, were examined in a derivative of the hormone-sensitive prostate cancer cell line LNCAP: The LNCaP TGF-beta receptor II cells, engineered to express TGF-beta receptor II, are sensitive to both DHT and TGF-beta. Surprisingly, when the LNCaP TGF-beta receptor II cells were treated with TGF-beta in the presence of physiological levels of DHT, both cell cycle arrest and apoptosis induction were significantly enhanced over TGF-beta alone. This effect temporally correlated with an increased expression of the cell cycle regulator p21 as well as the apoptotic executioner, procaspase-1, and a parallel down-regulation of the antiapoptotic protein, bcl-2. Expression of bax and caspase-3 proteins remained unchanged following treatment. Furthermore, apoptosis induction was suppressed by the caspase-1 inhibitor, z-YVAD, but not the caspase-3 inhibitor, z-DQMD, thus demonstrating the functional significance of increased procaspase-1 expression in TGF-beta-mediated apoptosis in prostate cancer cells. These results indicate that TGF-beta-mediated apoptosis can actually be enhanced by androgens through specific mechanisms involving cell cycle and apoptosis regulators and provide initial evidence on the ability of physiological levels of androgens to stimulate the intrinsic apoptotic potential of prostate cancer cells. Therefore, this study provides a molecular basis for the priming of prostate cancer cells for maximal apoptosis induction, during hormone- ablation therapy.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins , Dihydrotestosterone/pharmacology , Prostatic Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins , Blotting, Western , Caspase 3 , Caspase Inhibitors , Caspases/genetics , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Drug Interactions , Enzyme Inhibitors/pharmacology , Humans , Male , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/analysis , RNA, Messenger/analysis , Receptors, Transforming Growth Factor beta/analysis , Receptors, Transforming Growth Factor beta/physiology , Tumor Cells, Cultured
17.
J Urol ; 165(6 Pt 1): 2126-30, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11371940

ABSTRACT

PURPOSE: Radiation induced apoptosis of prostate cancer cells may have therapeutic and prognostic significance in patients treated with radiotherapy. We determined whether the ability of prostate tumor cells to undergo apoptosis has potential value for predicting the clinical response of patients with prostate cancer to brachytherapy. MATERIALS AND METHODS: A total of 76 patients with clinical stages T1 to 2 disease who were not receiving adjuvant therapy underwent transperineal implantation with 125iodine or 103palladium seeds and biopsy 7 to 23 months (median 12) after therapy. Nonresponders were classified using the American Society for Therapeutic Radiology and Oncology criteria. The apoptotic index was analyzed using the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling assay in archived biopsy specimens from 76 treated and 19 matched pretreatment control patients. Serial sections of prostatic tumors were also evaluated for the expression of bax and bcl-2 proteins (apoptosis regulators) by immunohistochemical testing. RESULTS: A significant increase in the apoptotic index was detected in post-brachytherapy compared with pretreatment prostate specimens (3.1% versus 2%, p <0.05), as well as in patients with negative biopsy at followup compared with those with persistent malignancy (3.4% versus 1.8%, p = 0.02). In addition, there was a significant elevation in bcl-2 expression in prostatic tissue in patients with treatment failure compared with responders (30.5% versus 13.1%, p <0.05). CONCLUSIONS: To our knowledge this is the first study to establish a correlation of apoptosis induction and bcl-2 over expression with treatment outcome in patients with prostate cancer after brachytherapy. Our findings have significant clinical implications for identifying the value of the apoptotic index and bcl-2 expression in prostatic tumors for predicting the therapeutic response to brachytherapy.


Subject(s)
Adenocarcinoma/physiopathology , Adenocarcinoma/radiotherapy , Apoptosis , Brachytherapy , Genes, bcl-2/physiology , Prostatic Neoplasms/physiopathology , Prostatic Neoplasms/radiotherapy , Adenocarcinoma/metabolism , Aged , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Middle Aged , Prostatic Neoplasms/metabolism , Retrospective Studies
18.
Cancer Res ; 61(3): 1227-32, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11221855

ABSTRACT

Activation of the caspase cascade is involved in the execution of apoptosis in a variety of cellular systems. Recent studies demonstrated that caspase-1 activation was required for human prostate cancer cells to undergo apoptosis in response to transforming growth factor-beta (Y. Guo and N. Kyprianou, Cancer Res., 59: 1366-1371, 1999). In the present study, to identify the significance of caspases in prostate cancer progression, we examined the expression of three key caspases, caspase-1, caspase-3, and caspase-9, in normal and malignant human prostates. Caspase-1, -3, and -9 expression was examined at the mRNA and the protein level in a series of human normal and malignant prostate specimens. No significant differences were observed in the mRNA expression in prostatic tumors relative to the normal gland for any of the three caspases. Immunohistochemical analysis revealed that the pattern of protein expression and distribution was uniformly homogeneous in the normal prostate, with the epithelial cells exhibiting a diffuse cytoplasmic staining for caspase-1 and caspase-3. Significantly, the majority of primary prostate cancer specimens (80%) had total lack of caspase-1 immunoreactivity, whereas the remaining showed a significantly reduced expression compared with the normal prostate (P < 0.05). Caspase-3 expression was also reduced in moderately and poorly differentiated prostatic tumors compared with well-differentiated prostate adenocarcinomas and the normal prostate (P < 0.05). No significant correlation was found between the apoptotic index or Gleason grade and the pattern of caspase protein expression in the primary prostatic tumors analyzed. Western blot analysis revealed constitutive expression of the proenzyme forms of caspase-1, -3, and -9 in the human prostate cancer cell lines PC-3, DU-145, TSU-Pr1m and LNCaP, but caspase-1 expression was low in the less tumorigenic cell lines, DU-145 and LNCaP. These findings implicate the loss of caspase-1 protein as a potential step in the loss of apoptotic control during prostate tumorigenesis. This study suggests that the pattern of caspase-1 and -3 expression in prostatic tumors may have prognostic significance in disease progression.


Subject(s)
Caspase 1/biosynthesis , Caspases/biosynthesis , Prostatic Neoplasms/enzymology , Blotting, Western , Caspase 1/genetics , Caspase 3 , Caspase 9 , Caspases/genetics , Humans , Linear Models , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
19.
Prostate ; 46(1): 45-51, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11170131

ABSTRACT

BACKGROUND: Medical treatment of benign prostatic hyperplasia (BPH) targets relief of symptoms by causing either relaxation of the prostatic smooth muscle with alpha1 adrenergic blockade, or shrinkage of the gland with 5alpha-reductase inhibitors. We recently demonstrated that alpha1-blockers, such as terazosin, induce apoptosis in prostatic cells. In this study, we examined the combined effect of finasteride and terazosin on the rate of apoptosis and cellular proliferation to investigate their potential synergy at the cellular level. METHODS: Prostate specimens were obtained from men who were treated with either finasteride (n = 24), terazosin (n = 42), or combination therapy (n = 10) for varying time periods (1 week to 36 months) for the relief of the symptoms of BPH. The proliferative and apoptotic indices of both stromal and epithelial prostatic cell populations were determined. Antibodies against TGF-beta1 and TbetaRII were used to examine the immunoreactivity of TGF-beta1 and TbetaRII, respectively, in all prostate tissue sections. RESULTS: The apoptotic index in both prostate cell populations was significantly higher following the combination treatment compared to terazosin or finasteride alone. There were no significant changes in the rate of cellular proliferation with any treatment. Furthermore, there was a significant increase in TGF-beta1 expression in the prostates of patients treated with terazosin or combination therapy, while there was no change in TbetaRII expression. CONCLUSIONS: These results support the concept that induction of prostate apoptosis is a potential molecular mechanism underlying the combination effect of alpha1 blockade with 5alpha-reductase inhibitors in the effective treatment of BPH. The upregulation of TGF-beta1 implies a role for this ligand as an effector of apoptosis induction in response to alpha1-blockade or finasteride therapy of BPH patients.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Enzyme Inhibitors/therapeutic use , Finasteride/therapeutic use , Prazosin/analogs & derivatives , Prazosin/therapeutic use , Prostatic Hyperplasia/drug therapy , Adrenergic alpha-Antagonists/administration & dosage , Antibodies, Monoclonal , Apoptosis/drug effects , Cell Division/drug effects , Drug Combinations , Enzyme Inhibitors/administration & dosage , Finasteride/administration & dosage , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Ki-67 Antigen/chemistry , Linear Models , Male , Prazosin/administration & dosage , Prostate/drug effects , Prostate/pathology , Receptors, Transforming Growth Factor beta/analysis , Regression Analysis , Retrospective Studies , Transforming Growth Factor beta/analysis
20.
Curr Opin Urol ; 11(1): 27-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11148743

ABSTRACT

The standard medical therapy for symptomatic benign prostatic hyperplasia is still alpha-blockers and 5alpha-reductase inhibitors. Ongoing studies demonstrate the long-term safety and efficacy of these two classes of therapeutic approaches. Although there have been no new Food and Drug Administration approved medical therapies for the treatment of benign prostatic hyperplasia over the past year, interest in and the use of phytotherapeutic agents continues to increase. In this review, we will discuss the developments that have occurred over the past year in the medical management of benign prostatic hyperplasia. In addition, we present ongoing efforts at our center to obtain a better understanding of and manipulate the apoptotic pathway as it pertains to the pathophysiology of benign prostatic hyperplasia.


Subject(s)
Prostatic Hyperplasia/drug therapy , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/therapeutic use , Adrenergic alpha-Antagonists/therapeutic use , Apoptosis/drug effects , Forecasting , Humans , Male , Phytotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...