Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Biomech ; 134: 110991, 2022 03.
Article in English | MEDLINE | ID: mdl-35176590

ABSTRACT

Obesity is a primary risk factor for osteoarthritis. While previous work has addressed relationships between in vivo cartilage mechanics, composition, and obesity in the tibiofemoral joint, there is limited information on these relationships in the patellofemoral joint. The purpose of this study was to compare the patellofemoral cartilage mechanical response to walking in participants with normal and obese body mass indices (BMIs). Additionally, patellar cartilage T1rho relaxation times were measured before exercise to characterize the biochemical composition of the tissue. Fifteen participants (eight with normal BMI and seven with obese BMI) underwent baseline magnetic resonance imaging (MRI) of their right knee. They then walked on a treadmill for 20 min at a speed normalized to their leg length before a second MRI scan. Subsequently, three-dimensional models of the bones and articular surfaces of the patellofemoral joint were created via manual segmentation of the pre- and post-exercise MR images to compute cartilage thickness and strain. Strain was defined as the change in patellofemoral cartilage thickness normalized to the baseline thickness. Results showed that participants with an obese BMI exhibited significantly increased patellofemoral cartilage strain compared to those with a normal BMI (5.4 ± 4% vs. 1.7 ± 3%, respectively; p = 0.003). Furthermore, patellar cartilage T1rho values were significantly higher in participants with obese versus normal BMIs (95 ms vs. 83 ms, respectively; p = 0.049), indicative of decreased proteoglycan content in those with an obese BMI. In summary, the altered patellofemoral cartilage strain and composition observed in those with an obese BMI may be indicative of cartilage degeneration.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Patellofemoral Joint , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/pathology , Humans , Knee Joint/pathology , Magnetic Resonance Imaging/methods , Obesity/complications , Obesity/diagnostic imaging , Osteoarthritis, Knee/pathology , Patella/diagnostic imaging , Patellofemoral Joint/diagnostic imaging , Patellofemoral Joint/pathology
2.
Oncogenesis ; 6(5): e342, 2017 May 29.
Article in English | MEDLINE | ID: mdl-28553931

ABSTRACT

Interleukin-4 plays a critical role in the regulation of immune responses and has been detected at high levels in the tumour microenvironment of cancer patients, where concentrations correlate with the grade of malignancy. In prostate cancer, interleukin-4 has been associated with activation of the androgen receptor, increased proliferation and activation of survival pathways such as Akt and NF-κB. However, its role in therapy resistance has not yet been determined. Here we investigate the influence of interleukin-4 on primary epithelial cells from prostate cancer patients. Our data demonstrate an increase in the clonogenic potential of these cells when cultured in the presence of interleukin-4. In addition, a Phospho-Kinase Array revealed that in contrast to previously published work, signal transducer and activator of transcription6 (STAT6) is the only signalling molecule activated after interleukin-4 treatment. Using the STAT6-specific inhibitor AS1517499 we could confirm the role of STAT6 in increasing colony-forming frequency. However, clonogenic recovery assays revealed that interleukin-4 does not rescue the effects of either irradiation or docetaxel treatment. We therefore propose that although the interleukin-4/STAT6 axis does not appear to be involved in therapy resistance, it does play a crucial role in the colony-forming abilities of the basal cell population in prostate cancer. IL-4 may therefore contribute to disease relapse by providing a niche that is favourable for the clonogenic growth of prostate cancer stem cells.

3.
Cell Death Differ ; 23(12): 1973-1984, 2016 12.
Article in English | MEDLINE | ID: mdl-27447114

ABSTRACT

Chromosomal abnormalities are implicated in a substantial number of human developmental syndromes, but for many such disorders little is known about the causative genes. The recently described 1q41q42 microdeletion syndrome is characterized by characteristic dysmorphic features, intellectual disability and brain morphological abnormalities, but the precise genetic basis for these abnormalities remains unknown. Here, our detailed analysis of the genetic abnormalities of 1q41q42 microdeletion cases identified TP53BP2, which encodes apoptosis-stimulating protein of p53 2 (ASPP2), as a candidate gene for brain abnormalities. Consistent with this, Trp53bp2-deficient mice show dilation of lateral ventricles resembling the phenotype of 1q41q42 microdeletion patients. Trp53bp2 deficiency causes 100% neonatal lethality in the C57BL/6 background associated with a high incidence of neural tube defects and a range of developmental abnormalities such as congenital heart defects, coloboma, microphthalmia, urogenital and craniofacial abnormalities. Interestingly, abnormalities show a high degree of overlap with 1q41q42 microdeletion-associated abnormalities. These findings identify TP53BP2 as a strong candidate causative gene for central nervous system (CNS) defects in 1q41q42 microdeletion syndrome, and open new avenues for investigation of the mechanisms underlying CNS abnormalities.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Chromosome Deletion , Tumor Suppressor Proteins/deficiency , Animals , Apoptosis Regulatory Proteins/metabolism , Brain/abnormalities , Brain/pathology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/pathology , Female , Gene Deletion , Heart Ventricles/abnormalities , Heart Ventricles/pathology , Magnetic Resonance Imaging , Mice, Inbred BALB C , Mice, Inbred C57BL , Neural Tube Defects/pathology , Phenotype , Syndrome , Tumor Suppressor Proteins/metabolism
4.
Cancer Res ; 75(11): 2326-36, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25858144

ABSTRACT

Altered microRNA (miRNA; miR) expression is associated with tumor formation and progression of various solid cancers. A major challenge in miRNA expression profiling of bulk tumors is represented by the heterogeneity of the subpopulations of cells that constitute the organ, as well as the tumor tissue. Here, we analyzed the expression of miRNAs in a subpopulation of epithelial stem/progenitor-like cells in human prostate cancer [prostate cancer stem cell (PCSC)] and compared their expression profile to more differentiated cancer cells. In both cell lines and clinical prostate cancer specimens, we identified that miR-25 expression in PCSCs was low/absent and steadily increased during their differentiation into cells with a luminal epithelial phenotype. Functional studies revealed that overexpression of miR-25 in prostate cancer cell lines and selected subpopulation of highly metastatic and tumorigenic cells (ALDH(high)) strongly affected the invasive cytoskeleton, causing reduced migration in vitro and metastasis via attenuation of extravasation in vivo. Here, we show, for the first time, that miR-25 can act as a tumor suppressor in highly metastatic PCSCs by direct functional interaction with the 3'-untranslated regions of proinvasive αv- and α6-integrins. Taken together, our observations suggest that miR-25 is a key regulator of invasiveness in human prostate cancer through its direct interactions with αv- and α6-integrin expression.


Subject(s)
Integrin alpha6/biosynthesis , Integrin alphaV/biosynthesis , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic/genetics , Humans , Integrin alpha6/genetics , Integrin alphaV/genetics , Male , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology
5.
J Electromyogr Kinesiol ; 24(4): 497-501, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24768278

ABSTRACT

Individuals with knee OA often exhibit greater co-contraction of antagonistic muscle groups surrounding the affected joint which may lead to increases in dynamic joint stiffness. These detrimental changes in the symptomatic limb may also exist in the contralateral limb, thus contributing to its risk of developing knee osteoarthritis. The purpose of this study is to investigate the interlimb symmetry of dynamic knee joint stiffness and muscular co-contraction in knee osteoarthritis. Muscular co-contraction and dynamic knee joint stiffness were assessed in 17 subjects with mild to moderate unilateral medial compartment knee osteoarthritis and 17 healthy control subjects while walking at a controlled speed (1.0m/s). Paired and independent t-tests determined whether significant differences exist between groups (p<0.05). There were no significant differences in dynamic joint stiffness or co-contraction between the OA symptomatic and OA contralateral group (p=0.247, p=0.874, respectively) or between the OA contralateral and healthy group (p=0.635, p=0.078, respectively). There was no significant difference in stiffness between the OA symptomatic and healthy group (p=0.600); however, there was a slight trend toward enhanced co-contraction in the symptomatic knees compared to the healthy group (p=0.051). Subjects with mild to moderate knee osteoarthritis maintain symmetric control strategies during gait.


Subject(s)
Muscle, Skeletal/physiopathology , Osteoarthritis, Knee/physiopathology , Walking/physiology , Aged , Case-Control Studies , Female , Gait/physiology , Humans , Knee Joint/physiopathology , Male , Middle Aged , Muscle Contraction/physiology
6.
Cell Death Differ ; 21(5): 761-73, 2014 May.
Article in English | MEDLINE | ID: mdl-24464224

ABSTRACT

Prostate cancer (CaP) is mostly composed of luminal-like differentiated cells, but contains a small subpopulation of basal cells (including stem-like cells), which can proliferate and differentiate into luminal-like cells. In cancers, CpG island hypermethylation has been associated with gene downregulation, but the causal relationship between the two phenomena is still debated. Here we clarify the origin and function of CpG island hypermethylation in CaP, in the context of a cancer cell hierarchy and epithelial differentiation, by analysis of separated basal and luminal cells from cancers. For a set of genes (including GSTP1) that are hypermethylated in CaP, gene downregulation is the result of cell differentiation and is not cancer specific. Hypermethylation is however seen in more differentiated cancer cells and is promoted by hyperproliferation. These genes are maintained as actively expressed and methylation-free in undifferentiated CaP cells, and their hypermethylation is not essential for either tumour development or expansion. We present evidence for the causes and the dynamics of CpG island hypermethylation in CaP, showing that, for a specific set of genes, promoter methylation is downstream of gene downregulation and is not a driver of gene repression, while gene repression is a result of tissue-specific differentiation.


Subject(s)
DNA Methylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Cell Differentiation/genetics , Cell Growth Processes/genetics , Down-Regulation , Epithelial Cells/pathology , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Prognosis , Tumor Cells, Cultured
7.
Br J Cancer ; 109(12): 3023-33, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24220693

ABSTRACT

BACKGROUND: Radiotherapy can be an effective treatment for prostate cancer, but radiorecurrent tumours do develop. Considering prostate cancer heterogeneity, we hypothesised that primitive stem-like cells may constitute the radiation-resistant fraction. METHODS: Primary cultures were derived from patients undergoing resection for prostate cancer or benign prostatic hyperplasia. After short-term culture, three populations of cells were sorted, reflecting the prostate epithelial hierarchy, namely stem-like cells (SCs, α2ß1integrin(hi)/CD133(+)), transit-amplifying (TA, α2ß1integrin(hi)/CD133(-)) and committed basal (CB, α2ß1integrin(lo)) cells. Radiosensitivity was measured by colony-forming efficiency (CFE) and DNA damage by comet assay and DNA damage foci quantification. Immunofluorescence and flow cytometry were used to measure heterochromatin. The HDAC (histone deacetylase) inhibitor Trichostatin A was used as a radiosensitiser. RESULTS: Stem-like cells had increased CFE post irradiation compared with the more differentiated cells (TA and CB). The SC population sustained fewer lethal double-strand breaks than either TA or CB cells, which correlated with SCs being less proliferative and having increased levels of heterochromatin. Finally, treatment with an HDAC inhibitor sensitised the SCs to radiation. INTERPRETATION: Prostate SCs are more radioresistant than more differentiated cell populations. We suggest that the primitive cells survive radiation therapy and that pre-treatment with HDAC inhibitors may sensitise this resistant fraction.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Aged , Aged, 80 and over , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Comet Assay , DNA Damage , Humans , Male , Middle Aged , Neoplastic Stem Cells/radiation effects , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
8.
Oncogenesis ; 2: e45, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23588494

ABSTRACT

The mouse haematopoietic stem cell (SC) regulator Latexin (LXN) is the only known homologue of the retinoic acid receptor responder 1 (RARRES1) gene. Both genes lie adjacent on chromosome 3 and differ mostly by the presence of a transmembrane domain in RARRES1. Despite their homology, it is not known whether they possess similar regulatory mechanisms, cellular localization and function. Here, we identified RARRES1 and LXN as highly significantly downregulated genes in human prostate SCs, whose expression was induced by the pro-differentiation agent all-trans retinoic acid (atRA). AtRA induced expression in the most differentiated cells compared with the SC fraction, suggesting that this subpopulation was less responsive to atRA. Small interfering RNA suppression of RARRES1 and LXN enhanced the SC properties of primary prostate cultures, as shown by a significant increase in their colony-forming ability. Expression of both RARRES1 and LXN was co-ordinately repressed by DNA methylation in prostate cancer cell lines and inhibition of RARRES1 and LXN increased the invasive capacity of primary prostate cultures, which also fully rescued an inhibitory effect induced by atRA. Moreover, we showed that RARRES1 and LXN reside within different sub-cellular compartments, providing evidence that RARRES1 is not a plasma membrane protein as previously supposed but is located primarily in the endoplasmic reticulum; whereas LXN was detected in the nucleus of prostate epithelial cells. Thus, LXN and RARRES1 are potential tumour suppressor genes, which are co-ordinately regulated, SC-silenced genes functioning to suppress invasion and colony-forming ability of prostate cancer cells; yet the proteins reside within different sub-cellular compartments.

9.
J Pathol ; 217(2): 299-306, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19040209

ABSTRACT

Despite the discovery over 60 years ago by Huggins and Hodges that prostate cancers respond to androgen deprivation therapy, hormone-refractory prostate cancer remains a major clinical challenge. There is now mounting evidence that solid tumours originate from undifferentiated stem cell-like cells coexisting within a heterogeneous tumour mass that drive tumour formation, maintain tumour homeostasis and initiate metastases. This review focuses upon current evidence for prostate cancer stem cells, addressing the identification and properties of both normal and transformed prostate stem cells.


Subject(s)
Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Animals , Cell Transformation, Neoplastic , Humans , Male , Models, Animal , Prostate/cytology , Stem Cell Niche , Stem Cells/cytology
10.
Article in English | MEDLINE | ID: mdl-17939301

ABSTRACT

Prostate cancer is now a common disease in men over 50 years of age. Medical therapies for prostate cancer are based on discoveries from the mid-twentieth century, and in the long term are rarely curative. Most treatments are directed towards an androgen receptor-expressing, highly proliferative target cell, which does indeed form the vast majority of cells in a prostate tumour. However, by invoking the existence of a cancer stem cell which, like normal epithelial stem cells in the prostate, does not express androgen receptor and is relatively quiescent, the observed resistance to most medical therapies can be explained. The phenotype of the prostate cancer stem cells is that of a basal cell and cultures derived from cancers, but not benign tissues, express a range of prostate cancer-associated RNAs. Furthermore, stem cells purified on the basis of alpha2beta1 high integrin and CD133 cell surface antigen expression, from an established culture of Gleason 4 (2+2) prostate cancer (P4E6), were able to form multiple intraprostatic tumours in nude mice when grafted orthotopically in a matrigel plug containing human prostatic stroma. The final tumours reexpressed androgen receptor and displayed a histology similar to that of a Gleason 4 cancer.


Subject(s)
Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Stem Cells/cytology , Cell Separation , Gene Expression , Genetic Therapy , Humans , Immunotherapy , Male , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/surgery
11.
Br J Cancer ; 92(12): 2171-80, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15928670

ABSTRACT

Extracellular proteases of the matrix metalloproteinase (MMP) and serine protease families participate in many aspects of tumour growth and metastasis. Using quantitative real-time RT-PCR analysis, we have undertaken a comprehensive survey of the expression of these enzymes and of their natural inhibitors in 44 cases of human prostate cancer and 23 benign prostate specimens. We found increased expression of MMP10, 15, 24, 25 and 26, urokinase plasminogen activator-receptor (uPAR) and plasminogen activator inhibitor-1 (PAI1), and the newly characterised serine proteases hepsin and matriptase-1 (MTSP1) in malignant tissue compared to benign prostate tissue. In contrast, there was significantly decreased expression of MMP2 and MMP23, maspin, and the protease inhibitors tissue inhibitor of metalloproteinase 3 (TIMP3), TIMP4 and RECK (reversion-inducing cysteine-rich protein with Kazal motifs) in the cancer specimens. The expression of MMP15 and MMP26 correlated positively with Gleason score, whereas TIMP3, TIMP4 and RECK expression correlated negatively with Gleason score. The cellular localisation of the expression of the deregulated genes was evaluated using primary malignant epithelial and stromal cell cultures derived from radical prostatectomy specimens. MMP10 and 25, hepsin, MTSP1 and maspin showed predominantly epithelial expression, whereas TIMP 3 and 4, RECK, MMP2 and 23, uPAR and PAI1 were produced primarily by stromal cells. These data provide the first comprehensive and quantitative analysis of the expression and localisation of MMPs and their inhibitors in human prostate cancer, leading to the identification of several genes involved in proteolysis as potential prognostic indicators, in particular hepsin, MTSP1, MMP26, PAI1, uPAR, MMP15, TIMP3, TIMP4, maspin and RECK.


Subject(s)
Matrix Metalloproteinases/genetics , Prostatic Neoplasms/genetics , Serine Endopeptidases/genetics , Tissue Inhibitor of Metalloproteinases/genetics , Aged , Disease Progression , GPI-Linked Proteins , Gene Expression Profiling , Humans , Male , Matrix Metalloproteinases/biosynthesis , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Middle Aged , Prognosis , Prostate/metabolism , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/biosynthesis , Tissue Inhibitor of Metalloproteinases/biosynthesis
12.
J Cell Sci ; 114(Pt 21): 3865-72, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11719553

ABSTRACT

A major impediment to our understanding of the biology of stem cells is the inability to distinguish them from their differentiating progeny. We made use of the known association of stem cells with basement membranes to isolate prostate epithelial stem cells. We show that, in vivo, putative stem cells express higher levels of the alpha(2)-integrin subunit than other cells within the basal layer. Approximately 1% of basal cells examined by confocal microscopy were integrin "bright", and these cells can be selected directly from the tissue on the basis of rapid adhesion to type I collagen. This selected population has a basal phenotype, as determined by expression of CK5 and CK14 and lack of expression of the differentiation-specific markers prostate specific antigen (PSA) and prostatic acid phosphatase (PAP), and has a fourfold greater ability to form colonies in vitro than the total basal population. These putative stem cells are distinguished from other basal cells by their ability to generate prostate-like glands in vivo with morphologic and immuno-histochemical evidence of prostate-specific differentiation. These properties are consistent with a stem cell origin. Furthermore, the presence of surface integrins on prostate stem cells suggests that these cells share common pathways with stem cells in other tissues.


Subject(s)
Integrins/metabolism , Prostate/metabolism , Stem Cells/metabolism , Aged , Aged, 80 and over , Cell Adhesion , Cell Differentiation , Cell Division , Cell Separation , Cells, Cultured , Collagen/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Male , Middle Aged , Prostate/cytology , Receptors, Collagen , Stem Cells/cytology
13.
J Endocrinol ; 160(2): 257-66, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9924195

ABSTRACT

Transforming growth factor-beta1 (TGFbeta1) is inhibitory to most epithelia, but its role in the control of proliferation of prostatic epithelium is unclear. In some cells, TGFbeta1 inhibition is achieved by up-regulation of cyclin-dependent kinase (cdk) inhibitors including p15, p21 and p27. Our aims were to determine whether the effects of TGFbeta1 on human prostatic epithelial cell cycle kinetics were mediated by alterations in the levels of the cdk inhibitors p15, p16, p21 and p27 and hypo-phosphorylated retinoblastoma protein (Rb). Human prostatic epithelial cells in primary culture were grown in the presence of TGFbeta1 (0-10 ng/ml) for up to 4 days and proliferation assessed using a [3H]thymidine uptake assay. Levels of p15, p16, p21 and p27 were measured at both mRNA and protein level by means of a reverse transcriptase PCR-based assay and Western analysis. Rb and cdk2 levels were measured. Exogenous TGFbeta1 (0-5 ng/ml) inhibited proliferation. This was associated with blocking of the cell cycle at G1, and up to 4-fold increases in p15, p21 and p27 mRNA levels, but no change was observed in p16 mRNA levels; these changes were not blocked by cycloheximide. Increased levels of p15, p21 and p27 protein were also accompanied by increased levels of hypo-phosphorylated Rb and decreased cdk2 kinase activity. TGFbeta1 has mainly inhibitory effects on benign human prostatic epithelium, which are caused by up-regulation of cdk inhibitors, hypo-phosphorylation of Rb and delaying of the cell cycle in G1.


Subject(s)
Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinases/antagonists & inhibitors , Prostate/drug effects , Prostatic Hyperplasia/pathology , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins , Analysis of Variance , Carrier Proteins/metabolism , Cell Cycle/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Epithelium/drug effects , Epithelium/pathology , Flow Cytometry , G1 Phase , Gene Expression Regulation , Humans , Male , Prostate/pathology , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Prostate ; 37(3): 149-60, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9792132

ABSTRACT

BACKGROUND: Human prostatic epithelium consists mainly of basal and secretory luminal cells: the origin of these phenotypes from a common stem cell, within the basal compartment, has been proposed but not yet demonstrated. METHODS: Analyses by light and electron microscopy, immunocytochemistry, and flow cytometry were used to determine lineage. The criteria for identifying the different phenotypes were characteristic morphology, and organization and expression of luminal- and basal-specific markers. RESULTS: After organoids attached, outgrowths appeared with cells maintaining close cell-to-cell associations. The dividing cell compartment contained a subpopulation of cells with stem-cell characteristics and a major population that may correspond to amplifying cells. The characteristics of the stem-cell phenotype included reactivity with antibodies CKbasal, CK14, and Ki67. The amplifying cells were characterized as an intermediate phenotype between basal and luminal, as reactivity was demonstrated with CKbasal, CK14, and CK18. As outgrowths eventually merged, multilayering was apparent and cells on the uppermost layer had numerous secretory vacuoles and reacted strongly with antibodies CK18 and CK19, androgen receptor, and prostate-specific antigen, which is characteristic of secretory luminal cells in vivo. In passaged cultures, loss of reactivity with CKbasal was detected; we postulate that this population contains the stem-cell fraction. CONCLUSIONS: These findings demonstrate that basal and luminal cells are of the same lineage and are derived from a common stem cell. Moreover, the progenitor stem cells reside within the basal compartment.


Subject(s)
Epithelial Cells/cytology , Prostate/cytology , Stem Cells/cytology , Cell Differentiation , Cell Separation , Cells, Cultured , Epithelial Cells/chemistry , Epithelial Cells/ultrastructure , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Keratins/analysis , Ki-67 Antigen/analysis , Male , Microscopy, Confocal , Microscopy, Electron , Prostate/chemistry , Prostate/ultrastructure , Prostate-Specific Antigen/analysis , Receptors, Androgen/analysis , Stem Cells/chemistry , Stem Cells/ultrastructure
15.
Prostate ; 32(4): 259-65, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9288184

ABSTRACT

BACKGROUND: Inhibitors of 5 alpha reductase (5 alpha R), the enzyme that converts testosterone to dihydrotestosterone (DHT), have been shown to retard the growth of hyperplastic prostates. This study evaluates the effects of the 5 alpha R inhibitor, epristeride, on cultured stromal and epithelial cells from benign, hyperplastic adult prostates. METHODS: [3H]-thymidine incorporation was used as a measure of proliferation. Prostate-specific antigen (PSA) was quantified by ELISA and reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS: Stromal cell proliferation in response to testosterone was dose-dependently inhibited by epristeride (1 x 10(-9) -3 x 10(-7) M, P < 0.05). However, epristeride had no effect on DHT-induced growth or the growth of androgen-unresponsive stroma. Upregulation of PSA secretion from epithelial cells by androgens was downregulated by epristeride (3 x 10(-9) M, P < 0.05) in testosterone-treated cells. Transforming growth factor beta-1 (TGF beta-1) secretion was downregulated by testosterone treatment and increased following treatment with epristeride (3 x 10(-9) M, P < 0.05). CONCLUSIONS: This demonstrates that epristeride specifically blocks testosterone-induced effects on prostatic cultures. TGF beta-1 may be a marker of 5 alpha reductase activity.


Subject(s)
5-alpha Reductase Inhibitors , Androstadienes/pharmacology , Enzyme Inhibitors/pharmacology , Prostate/drug effects , Prostatic Hyperplasia/pathology , Adult , Cell Division/drug effects , Cells, Cultured , Dihydrotestosterone/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Humans , Immunohistochemistry , Male , Polymerase Chain Reaction , Prostate/metabolism , Prostate/pathology , Prostate-Specific Antigen/analysis , Prostate-Specific Antigen/biosynthesis , Prostatectomy , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/surgery , RNA, Messenger/biosynthesis , Testosterone/pharmacology , Thymidine/metabolism , Transcription, Genetic/drug effects , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/biosynthesis
16.
Oncogene ; 15(9): 1115-20, 1997 Aug 28.
Article in English | MEDLINE | ID: mdl-9285567

ABSTRACT

Cellular interactions between stroma and epithelium are important in the growth and proliferation of prostate cancer. Peptide growth factors may facilitate the progression of prostate cancer as autocrine and/or paracrine factors. Keratinocyte Growth Factor (KGF or FGF7) has a differentiative and proliferative effect on the epithelium of the developing rat prostate. We investigated if KGF may act as a paracrine agent in human prostate cancer and examined the expression of KGF and Fibroblast Growth Factor Receptors (FGFRs) (IIIb and IIIc isoforms of the FGFR1 and FGFR2 genes). Sixty-five percent (11 out of 17 informative cases) of prostate cancers (CaP) expressed KGF mRNA by RT-PCR, while KGF expression was not detected in benign prostatic hyperplasia (BPH) (n = 6). Upregulation of KGF expression was related to hormone insensitive tumours (P<0.05). Tumour grade and stage were not associated with KGF expression. The source of KGF expression was further characterised using an in vitro primary culture model, showing its restriction to the prostatic stroma. The FGFR1IIIb isoform was expressed in all cases of prostate cancer (n = 17), and FGFR1IIIc mRNA was not detected. In the BPH group, FGFR1IIIb transcripts were detected in four out of six cases. FGFR2IIIb expression was detected in five of six cases of BPH and twelve out of seventeen (71%) cases of prostate cancer. In CaP, though not reaching statistical significance, the persistence of FGFR2IIIb expression appeared to be associated with hormone insensitive tumours (P=0.052). FGFR2IIIc expression was present in eleven of seventeen tumours but was absent in all six cases of BPH. Functional assessment of recombinant KGF in a proliferation assay demonstrated a mitogenic effect of up to 100% on cultured prostatic epithelial cells.


Subject(s)
Androgens/physiology , Fibroblast Growth Factors , Growth Substances/biosynthesis , Prostatic Neoplasms/metabolism , Animals , Cell Differentiation/drug effects , Epithelium/metabolism , Epithelium/pathology , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Growth Substances/pharmacology , Humans , Male , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
17.
J Endocrinol ; 151(2): 315-22, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8958793

ABSTRACT

The current study was undertaken, using cultures of prostatic epithelial and stromal cells, to determine the functional interactions between androgens, basic fibroblast growth factor (FGF2) and transforming growth factor-beta 1 (TGF beta 1) and their importance in maintaining stromal homeostasis. Treatment of stromal cells with TGF beta 1 significantly increased intracellular FGF2 and FGF2 sequestered to the extracellular matrix. FGF2 was also detected in stromal conditioned medium (SCM), but at levels 70-fold less than found in cell lysates. TGF beta 1 (0.1 ng/ml) treatment caused an initial increase of 86% in secreted FGF2 levels, but high concentrations of TGF beta 1 (5 ng/ml) decreased FGF2 levels by 38%, relative to the untreated control. Further studies showed that epithelial conditioned medium (ECM), androgen-treated, stromal conditioned medium (ASCM), but not SCM were mitogenic for stromal cells. Both ECM and ASCM caused a threefold increase in DNA synthesis. FGF2 may be the mediator of these interactions, since the mitogenic effect of both ECM and ASCM was significantly reduced by the addition of anti-FGF2 neutralising antibody. We hypothesise that the lack of response of stromal cells to SCM is due to TGF beta 1 blocking the mitogenic effect of FGF2. Thus down-regulation of TGF beta 1 synthesis, by androgens, results in stromal proliferation by ASCM.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Prostate/metabolism , Prostatic Hyperplasia/metabolism , Transforming Growth Factor beta/pharmacology , Cell Division/drug effects , Cell Separation , Cells, Cultured , DNA/biosynthesis , Epithelium/metabolism , Epithelium/pathology , Flow Cytometry , Humans , Male , Nandrolone/analogs & derivatives , Nandrolone/pharmacology , Prostate/pathology , Prostatic Hyperplasia/pathology , Testosterone Congeners/pharmacology , Transforming Growth Factor beta/biosynthesis
18.
J Endocrinol ; 143(2): 269-77, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7530286

ABSTRACT

Stromal cells derived from collagenase-digested benign hyperplastic adult prostates were isolated and grown in culture. Androgen and oestrogen receptor status were determined and growth in response to mibolerone (a synthetic androgen) and oestradiol-17 beta was measured. In addition, the ability of oestrogens to regulate the androgen receptor in stromal cells was investigated. [3H]Thymidine incorporation into DNA was stimulated by mibolerone in primary and secondary cultures, but sensitivity was lost with subsequent passages. Androgen stimulation of [3H]thymidine incorporation was consistently inhibited by the anti-androgen cyproterone acetate. Oestradiol-17 beta also stimulated [3H]thymidine incorporation into DNA, and this effect was inhibited by the anti-oestrogen tamoxifen. Sensitivity to oestradiol was lost with subsequent passages. A combination of mibolerone and oestradiol was not synergistic in increasing [3H]thymidine incorporation into DNA, but maximal stimulation occurred at 100-fold lower concentrations of mibolerone and oestradiol when the two hormones were applied in combination. Specific high-affinity [3H]mibolerone- and [3H]oestradiol-binding sites were demonstrated by radioligand binding in intact cells. The affinity for oestradiol binding to its receptor exceeded that quantified for mibolerone binding to the androgen receptor, whilst the number of oestradiol-binding sites was approximately tenfold less than that quantified for mibolerone. Treatment with oestradiol down-regulated the number of [3H]mibolerone binding sites 1.7-fold (P < 0.005) as early as day 2 after oestradiol treatment. In conclusion, we successfully cultured stromal cells derived from hyperplastic prostates which retained sensitivity to androgen and oestrogen.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Estradiol/pharmacology , Nandrolone/analogs & derivatives , Prostate/drug effects , Prostatic Hyperplasia/metabolism , Receptors, Androgen/metabolism , Testosterone Congeners/pharmacology , Cells, Cultured , Humans , Male , Nandrolone/pharmacology , Prostate/metabolism , Prostate/pathology , Protein Binding , Stimulation, Chemical , Tamoxifen/pharmacology
19.
Neurology ; 30(12): 1316-9, 1980 Dec.
Article in English | MEDLINE | ID: mdl-6450333

ABSTRACT

A 22-month-old child suffered accidental strangulation, which rendered him comatose with intermittent generalized tonic-clonic seizures. His elecroencephalogram (EEG) displayed widespread activity of alpha frequency unreactive to sensory stimuli. Upon clinical recovery, a slower posterior EEG rhythm, attenuated by eye opening, was detected, which was more consistent with the patient's age. This observation is remarkable, because of the rarity of reports of an alpha pattern after cerebral anoxia in young children and the subsequent EEG and clinical evolutions.


Subject(s)
Alpha Rhythm , Hypoxia, Brain/physiopathology , Neck Injuries , Adult , Coma/physiopathology , Down Syndrome/complications , Down Syndrome/physiopathology , Humans , Hypoxia, Brain/complications , Male , Seizures/physiopathology
20.
J Comput Assist Tomogr ; 4(3): 326-9, 1980 Jun.
Article in English | MEDLINE | ID: mdl-7372865

ABSTRACT

This report deals with the contribution of computed tomography (CT) to the diagnosis of congenital toxoplasmosis in two infants. Computed tomography revealed diffuse hydrocephalus and confirmed the periventricular nature of the brain calcifications. The clinical, radiographic, and CT findings are discussed.


Subject(s)
Brain Diseases/diagnostic imaging , Tomography, X-Ray Computed , Toxoplasmosis, Congenital/diagnostic imaging , Brain Diseases/parasitology , Humans , Infant , Infant, Newborn , Male , Skull/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...