151
|
Deeble PD, Murphy DJ, Parsons SJ, Cox ME. Interleukin-6- and cyclic AMP-mediated signaling potentiates neuroendocrine differentiation of LNCaP prostate tumor cells. Mol Cell Biol 2001; 21:8471-82. [PMID: 11713282 PMCID: PMC100010 DOI: 10.1128/mcb.21.24.8471-8482.2001] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neuroendocrine (NE) differentiation in prostatic adenocarcinomas has been reported to be an early marker for development of androgen independence. Secretion of mitogenic peptides from nondividing NE cells is thought to contribute to a more aggressive disease by promoting the proliferation of surrounding tumor cells. We undertook studies to determine whether the prostate cancer cell line LNCaP could be induced to acquire NE characteristics by treatment with agents that are found in the complex environment in which progression of prostate cancer towards androgen independence occurs. We found that cotreatment of LNCaP cells with agents that signal through cyclic AMP-dependent protein kinase (PKA), such as epinephrine and forskolin, and with the cytokine interleukin-6 (IL-6) promoted the acquisition of an NE morphological phenotype above that seen with single agents. Convergent IL-6 and PKA signaling also resulted in potentiated mitogen-activated protein kinase (MAPK) activation without affecting the level of signal transducer and activator of transcription or PKA activation observed with these agents alone. Cotreatment with epinephrine and IL-6 synergistically increased c-fos transcription as well as transcription from the beta4 nicotinic acetylcholine receptor subunit promoter. Potentiated transcription from these elements was shown to be dependent on the MAPK pathway. Most importantly, cotreatment with PKA activators and IL-6 resulted in increased secretion of mitogenic neuropeptides. These results indicate that PKA and IL-6 signaling participates in gene transcriptional changes that reflect acquisition of an NE phenotype by LNCaP cells and suggest that similar signaling mechanisms, particularly at sites of metastasis, may be responsible for the increased NE content of many advanced prostate carcinomas.
Collapse
Affiliation(s)
- P D Deeble
- Department of Microbiology and Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | | | | |
Collapse
|
152
|
Lee LF, Guan J, Qiu Y, Kung HJ. Neuropeptide-induced androgen independence in prostate cancer cells: roles of nonreceptor tyrosine kinases Etk/Bmx, Src, and focal adhesion kinase. Mol Cell Biol 2001; 21:8385-97. [PMID: 11713275 PMCID: PMC100003 DOI: 10.1128/mcb.21.24.8385-8397.2001] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The bombesin/gastrin-releasing peptide (GRP) family of neuropeptides has been implicated in various in vitro and in vivo models of human malignancies including prostate cancers. It was previously shown that bombesin and/or neurotensin (NT) acts as a survival and migratory factor(s) for androgen-independent prostate cancers. However, a role in the transition from an androgen-dependent to -refractory state has not been addressed. In this study, we investigate the biological effects and signal pathways of bombesin and NT on LNCaP, a prostate cancer cell line which requires androgen for growth. We show that both neurotrophic factors can induce LNCaP growth in the absence of androgen. Concurrent transactivation of reporter genes driven by the prostate-specific antigen promoter or a promoter carrying an androgen-responsive element (ARE) indicate that growth stimulation is accompanied by androgen receptor (AR) activation. Furthermore, neurotrophic factor-induced gene activation was also present in PC3 cells transfected with the AR but not in the parental line which lacks the AR. Given that bombesin does not directly bind to the AR and is known to engage a G-protein-coupled receptor, we investigated downstream signaling events that could possibly interact with the AR pathway. We found that three nonreceptor tyrosine kinases, focal adhesion kinase (FAK), Src, and Etk/BMX play important parts in this process. Etk/Bmx activation requires FAK and Src and is critical for neurotrophic factor-induced growth, as LNCaP cells transfected with a dominant-negative Etk/BMX fail to respond to bombesin. Etk's activation requires FAK, Src, but not phosphatidylinositol 3-kinase. Likewise, bombesin-induced AR activation is inhibited by the dominant-negative mutant of either Src or FAK. Thus, in addition to defining a new G-protein pathway, this report makes the following points regarding prostate cancer. (i) Neurotrophic factors can activate the AR, thus circumventing the normal growth inhibition caused by androgen ablation. (ii) Tyrosine kinases are involved in neurotrophic factor-mediated AR activation and, as such, may serve as targets of future therapeutics, to be used in conjunction with current antihormone and antineuropeptide therapies.
Collapse
Affiliation(s)
- L F Lee
- Department of Biological Chemistry and Cancer Center, University of California at Davis, 4645 2nd Ave., Sacramento, CA 95817, USA
| | | | | | | |
Collapse
|
153
|
Juarranz MG, Bolaños O, Gutiérrez-Cañas I, Lerner EA, Robberecht P, Carmena MJ, Prieto JC, Rodríguez-Henche N. Neuroendocrine differentiation of the LNCaP prostate cancer cell line maintains the expression and function of VIP and PACAP receptors. Cell Signal 2001; 13:887-94. [PMID: 11728828 DOI: 10.1016/s0898-6568(01)00199-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The molecular mechanisms involved in differentiation of prostate cancer cells to a neuroendocrine (NE) cell phenotype are not well understood. Here we used the androgen-dependent human prostate cancer cell line LNCaP to perform a systematic and broad analysis of the expression, pharmacology, and functionality of vasoactive intestinal peptide (VIP)/pituitary adenylate cyclase-activating peptide (PACAP) receptors. Reverse transcription polymerase chain reaction experiments, together with pharmacological approaches with a set of specific agonists and antagonists, demonstrated the presence of the three VIP/PACAP receptor subtypes (PAC1, VPAC1, and VPAC2 with a major role for VPAC1, acting through adenylate cyclase (AC) stimulation. An essentially similar pattern was observed by NE differentiated cells (4 days after serum deprivation) in spite of the important morphological changes observed. However, the expression of the prostate-specific antigen (PSA) decreased in NE cells (and increased again by dihydrotestosterone, DHT, treatment). The present demonstration of the induction of NE transdifferentiation in LNCaP cells by increasing concentrations of VIP adds value to previous observations on the role of cAMP in this process, an interesting topic in the comprehension of the molecular changes that are involved in the progression of prostate cancer to androgen independence.
Collapse
MESH Headings
- Adenylyl Cyclases/metabolism
- Binding, Competitive
- Cell Differentiation
- Culture Media, Serum-Free
- Cyclic AMP/biosynthesis
- Dose-Response Relationship, Drug
- Humans
- Male
- Neurites/ultrastructure
- Neurons/cytology
- Neurons/metabolism
- Neuropeptides/pharmacology
- Neurosecretory Systems/cytology
- Neurosecretory Systems/metabolism
- Pituitary Adenylate Cyclase-Activating Polypeptide
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA, Neoplasm/biosynthesis
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I
- Receptors, Pituitary Hormone/genetics
- Receptors, Pituitary Hormone/metabolism
- Receptors, Pituitary Hormone/physiology
- Receptors, Vasoactive Intestinal Peptide/genetics
- Receptors, Vasoactive Intestinal Peptide/metabolism
- Receptors, Vasoactive Intestinal Peptide/physiology
- Receptors, Vasoactive Intestinal Peptide, Type II
- Receptors, Vasoactive Intestinal Polypeptide, Type I
- Transcription, Genetic
- Tumor Cells, Cultured
- Vasoactive Intestinal Peptide/pharmacology
Collapse
Affiliation(s)
- M G Juarranz
- Department of Biochemistry and Molecular Biology, Universidad de Alcalá, 28871, Alcalá de Henares, Spain
| | | | | | | | | | | | | | | |
Collapse
|
154
|
van Leenders GJ, Aalders TW, Hulsbergen-van de Kaa CA, Ruiter DJ, Schalken JA. Expression of basal cell keratins in human prostate cancer metastases and cell lines. J Pathol 2001; 195:563-70. [PMID: 11745692 DOI: 10.1002/path.993] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Within normal human prostate epithelium, basal and luminal cells can be discriminated by their expression of keratins (K). While basal cells express K5/14, luminal cells show expression of K8/18 and an intermediate cell population can be identified by co-expression of K5/18. Prostate cancer is predominantly composed of luminal and neuroendocrine cells, while a minority of cells have a basal phenotype. In order to distinguish between basal and intermediate cells, and to assess the effects of androgen deprivation on prostate cancer, 56 human prostate cancer metastases and three cancer cell lines were characterized using antibodies to K5, K14, K18, and the neuroendocrine marker chromogranin A (ChA). The staining was performed on paraffin tissue and visualized by the avidin-biotin-peroxidase complex method. Protein expression was quantified as the number of positive cells in 20 high power fields (HPF; 400x). Keratin expression in the prostate cancer cell lines LNCaP, DU145, and PC3 was analysed by immunofluorescence with triple staining and confocal laser scanning microscopy. Prostate cancer metastases were consistently positive for K18 and negative for K14, irrespective of hormonal therapy. K5 expression was displayed in 28.9% of the tumours without treatment, in 75% after androgen deprivation, and in 57.1% of hormone-escaped prostate carcinomas. After androgen deprivation, the number of K5-expressing cells increased significantly. While androgen-dependent prostate cancer showed a median of 0 cells/20 HPF (range 0-50), regressed tumours displayed 22.5 (range 0-65) and hormone-escaped tumours 7.5 (range 0-361) positive cells/20 HPF. Expression of ChA was observed in 47.4% of the androgen-dependent tumours. The number of neuroendocrine cells was not significantly affected in regressed or hormone-escaped disease. The androgen-dependent cell line LNCaP stained for K18, while the androgen-independent lines DU145 and PC3 both expressed K5 and 18. Expression of K5 in the absence of K14 identifies the existence of an intermediate cell population in prostate carcinoma. Accumulation of intermediate cells in regressed and hormone-escaped prostate cancer indicates that for their survival, these cells are androgen-independent.
Collapse
Affiliation(s)
- G J van Leenders
- Department of Pathology, University Medical Centre St. Radboud, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
155
|
Wilson EM, Oh Y, Hwa V, Rosenfeld RG. Interaction of IGF-binding protein-related protein 1 with a novel protein, neuroendocrine differentiation factor, results in neuroendocrine differentiation of prostate cancer cells. J Clin Endocrinol Metab 2001; 86:4504-11. [PMID: 11549700 DOI: 10.1210/jcem.86.9.7845] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Neuroendocrine cells have been implicated in many cancers, including small cell lung, cervical, breast, and prostate carcinomas. The increase in neuroendocrine cell number in prostate cancer has been reported to correlate with poor prognosis, progressive tumors, and androgen insensitivity. The mechanisms involved in this differentiation remain unknown. IGF-binding protein-related protein 1 is a member of the IGF-binding protein superfamily and has recently been shown to exhibit differentiation and tumor suppression activity in prostate cancer cell lines stably overexpressing IGF-binding protein-related protein 1. From a yeast two-hybrid screen, a novel IGF-binding protein-related protein 1-interacting protein was identified. Immunocytochemical techniques indicate that this protein, 25.1, and intracellular IGF-binding protein-related protein 1 colocalize in the nucleus. When 25.1 is transiently expressed in a stable prostate cancer cell line overexpressing IGF-binding protein-related protein 1, cells assume a neuritic-like morphology with long dendritic-like processes and express the neuroendocrine markers chromogranin A and neuron-specific enolase. We propose that 25.1 (neuroendocrine differentiation factor) together with IGF-binding protein-related protein 1 can induce neuroendocrine cell differentiation in prostate cancer cells.
Collapse
Affiliation(s)
- E M Wilson
- Department of Pediatrics, Oregon Health Sciences University, Portland, Oregon 97201, USA
| | | | | | | |
Collapse
|
156
|
van Leenders GJ, Schalken JA. Stem cell differentiation within the human prostate epithelium: implications for prostate carcinogenesis. BJU Int 2001; 88 Suppl 2:35-42; discussion 49-50. [PMID: 11589668 DOI: 10.1046/j.1464-410x.2001.00117.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- G J van Leenders
- Department of Pathology, University Medical Centre St. Radboud, Nijmegen, The Netherlands.
| | | |
Collapse
|
157
|
Zermann DH, Ishigooka M, Schmidt RA. A bridge between neuroscience and oncology: prostate cancer genesis. A 'negative' consequence of learning? Med Hypotheses 2001; 57:201-6. [PMID: 11461173 DOI: 10.1054/mehy.2000.1294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Prostate cancer has become the most commonly diagnosed cancer in men over recent years. The initiating mechanism for tumorigenesis within the prostate remains an unknown. The observation, that the cancer incidence in patients with chronic neurological disabilities is significantly lower than in the normal population lead to the hypothesis, that changed processing due to a barrage of aberrant sensory information within a healthy CNS can trigger events within the prostate cell, that cause malignant transformation. There is a broad overlap of cellular mechanism of gene expression, that lead to either long term potentiation, learning and memory storage or deregulated differentiation and malignant transformation.
Collapse
Affiliation(s)
- D H Zermann
- Neuro-Urology Unit, Department of Urology, University of Colorado Health Science Center, Denver, CO, USA.
| | | | | |
Collapse
|
158
|
Zelivianski S, Verni M, Moore C, Kondrikov D, Taylor R, Lin MF. Multipathways for transdifferentiation of human prostate cancer cells into neuroendocrine-like phenotype. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1539:28-43. [PMID: 11389966 DOI: 10.1016/s0167-4889(01)00087-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The neuroendocrine (NE) cell is a minor cell population in normal human prostate glands. The number of NE cells is increased in advanced hormone-refractory prostate carcinomas (PCA). The mechanism of increased NE cell population in these advanced tumors is poorly understood. We examined molecular mechanisms which may be involved in the regulation of the transdifferentiation process of human PCA cells leading to a NE phenotype. We compared PCA cell lines LNCaP and PC-3 in the following medium conditions: steroid-reduced (SR), interleukin-6 (IL-6)-supplemented, or dibutyrate cAMP (db-cAMP)-supplemented. We found that androgen-responsive C-33 LNCaP cells responded to all treatments, having a neuronal-like morphology. In contrast, C-81 LNCaP cells, having a decreased androgen responsiveness, had a less pronounced effect although followed a similar trend. Androgen-unresponsive PC-3 cells showed little change in their morphology. Grown in the SR condition, the level of neuron-specific enolase (NSE), a marker of neuronal cells, was upregulated in C-33 LNCaP cells, while to a lesser degree in the presence of IL-6. In the presence of db-cAMP, the NSE level in C-33 cells was decreased, lower than that in control cells. An opposite effect was observed for C-81 LNCaP cells. Nevertheless, the NSE level was only elevated in db-cAMP-treated PC-3 cells, but no change was found in PC-3 cells grown in the SR- or IL-6-supplemented medium. Thus, a similar gross phenotypic change may correlate with differential molecular expressions. We also analyzed the expression of protein tyrosine phosphatase alpha (RPTPalpha) since it plays a critical role in normal neuronal differentiation and signaling. Our results showed that the expression of RPTPalpha correlates with the NE phenotypic change of LNCaP cells in the SR condition. In summary, our data clearly show that the molecular process by which cultured human prostate cancer cells undergo a transdifferentiation process to a NE cell-like phenotype is accompanied by differential expressions of different markers, and a gross NE cell-like phenotype can occur by exposing PCA cells to different pharmacological agents.
Collapse
Affiliation(s)
- S Zelivianski
- Department of Biochemistry/Molecular Biology, University of Nebraska Medical Center, Omaha 68198, USA
| | | | | | | | | | | |
Collapse
|
159
|
Deng W, Obrocka M, Fischer I, Prockop DJ. In vitro differentiation of human marrow stromal cells into early progenitors of neural cells by conditions that increase intracellular cyclic AMP. Biochem Biophys Res Commun 2001; 282:148-52. [PMID: 11263984 DOI: 10.1006/bbrc.2001.4570] [Citation(s) in RCA: 351] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Human marrow stromal cells (hMSCs) are multipotential stem cells that can be differentiated into bone, cartilage, fat, and muscle. In the experiments here, we found that undifferentiated cultures of hMSCs express some markers characteristic of neural cells such as microtubule-associated protein 1B (MAP1B), neuron-specific tubulin (TuJ-1), neuron-specific enolase (NSE), and vimentin. By treating hMSCs with 0.5 mM isobutylmethylxanthine (IBMX)/1 mM dibutyryl cyclic AMP (dbcAMP) for 6 days, about 25% of the hMSCs differentiated into cells with a typical neural cell morphology and with increased levels of both NSE and vimentin. The data suggested that the hMSCs may have been differentiated into early progenitors of neural cells in vitro under conditions that increase the intracellular level of cAMP.
Collapse
Affiliation(s)
- W Deng
- Center for Gene Therapy, Tulane University Health Sciences Center, SL-99, 1430 Tulane Avenue, New Orleans, Louisiana, 70112, USA
| | | | | | | |
Collapse
|
160
|
Aumüller G, Leonhardt M, Renneberg H, von Rahden B, Bjartell A, Abrahamsson PA. Semiquantitative morphology of human prostatic development and regional distribution of prostatic neuroendocrine cells. Prostate 2001; 46:108-15. [PMID: 11170138 DOI: 10.1002/1097-0045(20010201)46:2<108::aid-pros1014>3.0.co;2-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND The neuroendocrine cells of the human prostate have been related to proliferative disorders such as prostatic cancer. Their origin, distribution, and development have therefore been studied and discussed in terms of current stem cell concepts in the prostate. METHODS Prostatic tissue specimens (n = 20) from human fetuses (n = 8), prepubertal and pubertal children (n = 8) and mature men (n = 4) were studied immunohistochemically using antibodies directed against neuroendocrine, epithelial as well as secretory markers. Semiquantitative computer-assisted evaluation of different epithelial and stromal components based on stereological principles was performed on azan-stained sections representative of all developmental stages. RESULTS By the end of gestational Week 9, neuroendocrine (NE) cells appear in the epithelium of the urogenital sinus and are subsequently closely associated with the formation of urethral prostatic buds. The fetal and postnatal distribution pattern of NE cells within the gland is characterized by a relatively constant number of cells per gland similar to prostatic smooth muscle cells. Likewise, a density gradient exists with the highest density in the large collicular ducts and almost no NE cells in subcapsular peripheral acini. In peripheral ducts, the distribution is random. Maturation of the NE cells precedes that of the secretory cells by about 10-16 years. CONCLUSIONS A second prostatic stem cell lineage, different from the urogenital sinus (UGS)-lineage is hypothesized originating from immature neuroendocrine cells. Being morphologically indistinguishable from the UGS-derived prostatic secretory cell lineage, it gives rise to neuroendocrine cells. Their presence is apparently important for proliferation regulation of the UGS-derived lineage of the prostate.
Collapse
Affiliation(s)
- G Aumüller
- Department of Anatomy and Cell Biology, University of Marburg, D-35033 Marburg, Germany.
| | | | | | | | | | | |
Collapse
|
161
|
Clejan S, O'Connor K, Rosensweig N. Tri-dimensional prostate cell cultures in simulated microgravity and induced changes in lipid second messengers and signal transduction. J Cell Mol Med 2001; 5:60-73. [PMID: 12067451 PMCID: PMC6737775 DOI: 10.1111/j.1582-4934.2001.tb00138.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The high aspect rotating-wall vessel (HARV) was designed to cultivate cells in an environment that simulate microgravity. We studied previously the effects of HARV cultivation on DU-145 human prostate carcinoma cells. We determined that HARV cultivation produced a less aggressive, slower growing, less proliferative, more differentiated and less pliant cell than other cell cultivation methods. The result was a 3-dimensional (3D) growth model of prostate cancer which mimics in vivo tissue growth. This work examines the signal transduction-second messenger pathways existing temporarily in these HARV cells and correlates these features with the special properties in growth and 3D spheroid formation. We found an initial very active ceramide, a diacylglycerol increase together with increases in PI-PLC and PLA(2) a central defect in PLD (no phosphatic acid or phosphatidylethanol at any time during 15 days of HARV cultivation). There is a cross-talk between ceramide and PI3K pathways with activation of PI3K, after 6 days of HARV growth concomitant with down-regulation of ceramide. At this time, there is also an increase of cAMP (seen by increases in arachidonic acid). Taken together these results can explain the 3D organoid-like growth. We therefore developed a model for growth in HARV prostate cancer cells which involve temporal "switches" between second messengers, activation and cross-talk between multiplicity of signaling pathways and a central defect in PLD pathways. Essential to the late slow growth, and 3D organotypic formation are the apoptotic, anti-survival, anti-proliferation and differentiation pathways in the first days of HARV, with growth of "new" different types of prostate cancer cells which set-up for later "switch" in ceramide-PI3K to survival and proliferation.
Collapse
Affiliation(s)
- S Clejan
- Department of Pathology & Laboratory Medicine, Tulane University Health Science Center, 1430 Tulane Ave., SL 79, New Orleans, LA 70112-2699, USA.
| | | | | |
Collapse
|
162
|
Abstract
Previous studies have shown that calcitonin-like immunoreactive substances are secreted by primary prostate cells. Furthermore, exogenously added calcitonin stimulates proliferation of androgen-responsive LnCaP cells. To examine the possible effect of calcitonin on growth of invasive prostate cancer cells, we tested its effects on proliferation of PC-3M cells. Calcitonin stimulated DNA synthesis of PC-3M cells in a dose-dependent fashion, and also stimulated adenylyl cyclase and protein kinase C activities. To further delineate the role of these signaling cascades in proliferation of PC-3M prostate cancer cells, we selectively activated these pathways by transfecting cDNAs expressing constitutively active forms of either Gsalpha (Gsalpha-QL) or Gqalpha (Gqalpha-QL). cDNAs expressing wild-type forms of G-proteins (Gsalpha-WT and Gqalpha-WT) were used as vehicle controls. Gqalpha-QL transfectants exhibited growth inhibition and terminal differentiation. Those expressing Gsalpha-QL exhibited a dramatic increase in growth rate. Gsalpha-QL transfectants displayed an almost 3-fold increase in [3H]-thymidine incorporation and over a 4-fold increase in growth rate when compared with parental PC-3M cells or those expressing wild-type Gsalpha (Gsalpha-WT). The growth-promoting action of Gsalpha-QL could not be mimicked by either 8-bromo cAMP or forskolin. However, nifedipine, a calcium channel antagonist, potently and selectively inhibited DNA synthesis in Gsalpha-QL transfectants. These results suggest that the growth-promoting actions of Gsalpha on PC-3M cells may be mediated by nifedipine-sensitive proliferative events.
Collapse
Affiliation(s)
- J Chien
- Department of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, USA
| | | |
Collapse
|
163
|
Kim JS, Pirnia F, Choi YH, Nguyen PM, Knepper B, Tsokos M, Schulte TW, Birrer MJ, Blagosklonny MV, Schaefer O, Mushinski JF, Trepel JB. Lovastatin induces apoptosis in a primitive neuroectodermal tumor cell line in association with RB down-regulation and loss of the G1 checkpoint. Oncogene 2000; 19:6082-90. [PMID: 11146561 DOI: 10.1038/sj.onc.1204008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To develop a new approach to the treatment of primitive neuroectodermal tumors we evaluated the effect of the HMG-CoA reductase inhibitor lovastatin on the Ewing's sarcoma cell line CHP-100. Lovastatin induced neural morphology and markers including neuron-specific enolase and neurofilament protein. The acquisition of neural morphology required new mRNA synthesis, and cDNA microarray analysis confirmed that lovastatin altered the program of gene expression. After morphologic differentiation the cells underwent rapidly progressive apoptosis. In normal development of neuronal progenitors, differentiation signals trigger p21WAF1 accumulation, RB hypophosphorylation, enhanced RB-E2F-1 association, and G1 arrest, and these events have been shown to protect from apoptosis. In contrast, in the Ewing's sarcoma cells lovastatin triggered differentiation without causing cell cycle arrest: p21WAF1 was not induced, RB remained hyperphosphorylated, and RB protein expression and RB-E2F-1 association were markedly downregulated, suggesting that loss of an RB-regulated G1 checkpoint promoted apoptosis. Consistent with this hypothesis, adenoviral p21WAF1 decreased DNA synthesis and partially protected from lovastatin-induced cytotoxicity. The data demonstrate a new model for examining the genetic regulation of cell fate in a neural progenitor tumor and suggest a new approach to the treatment of this neoplasm.
Collapse
Affiliation(s)
- J S Kim
- Medicine Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Yuasa K, Kotera J, Fujishige K, Michibata H, Sasaki T, Omori K. Isolation and characterization of two novel phosphodiesterase PDE11A variants showing unique structure and tissue-specific expression. J Biol Chem 2000; 275:31469-79. [PMID: 10906126 DOI: 10.1074/jbc.m003041200] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
cDNAs encoding a novel phosphodiesterase, phosphodiesterase 11A (PDE11A), were isolated by a combination of reverse transcriptase-polymerase chain reaction using degenerate oligonucleotide primers and rapid amplification of cDNA ends. Their catalytic domain was identical to that of PDE11A1 (490 amino acids) reported during the course of this study. However, the cDNAs we isolated had N termini distinct from PDE11A1, indicating two novel N-terminal variants of PDE11A. PDE11A3 cDNA encoded a 684-amino acid protein including one complete and one incomplete GAF domain in the N-terminal region. PDE11A4 was composed of 934 amino acids including two complete GAF domains and shared 630 C-terminal amino acids with PDE11A3 but had a distinct N terminus containing the putative phosphorylation sites for cAMP- and cGMP-dependent protein kinases. PDE11A3 transcripts were specifically expressed in testis, whereas PDE11A4 transcripts were particularly abundant in prostate. Recombinant PDE11A4 expressed in COS-7 cells hydrolyzed cAMP and cGMP with K(m) values of 3.0 and 1.4 microm, respectively, and the V(max) value with cAMP was almost twice that with cGMP. Although PDE11A3 showed the same K(m) values as PDE11A4, the relative V(max) values of PDE11A3 were approximately one-sixth of those of PDE11A4. PDE11A4, but not PDE11A3, was phosphorylated by both cAMP- and cGMP-dependent protein kinases in vitro. Thus, the PDE11A gene undergoes tissue-specific alternative splicing that generates structurally and functionally distinct gene products.
Collapse
MESH Headings
- 3',5'-Cyclic-GMP Phosphodiesterases
- Alternative Splicing
- Amino Acid Sequence
- Amino Acids/chemistry
- Animals
- Base Sequence
- Blotting, Northern
- Blotting, Southern
- COS Cells
- Catalytic Domain
- Cloning, Molecular
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Cyclic GMP-Dependent Protein Kinases/metabolism
- DNA, Complementary/metabolism
- Databases, Factual
- Humans
- Hydrolysis
- Immunoblotting
- Inhibitory Concentration 50
- Kinetics
- Models, Genetic
- Molecular Sequence Data
- Nucleotides/metabolism
- Phosphoric Diester Hydrolases/biosynthesis
- Phosphoric Diester Hydrolases/chemistry
- Phosphoric Diester Hydrolases/genetics
- Phosphorylation
- Phosphotransferases/metabolism
- Plasmids/metabolism
- Precipitin Tests
- Protein Structure, Tertiary
- RNA Splicing
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Tissue Distribution
- Transfection
Collapse
Affiliation(s)
- K Yuasa
- Discovery Research Laboratory, Tanabe Seiyaku Co. Ltd., 2-50, Kawagishi-2-chome, Toda, Saitama 335-8505, Japan
| | | | | | | | | | | |
Collapse
|
165
|
Abstract
BACKGROUND Despite the beneficial effects of androgen ablation therapy in patients with prostate carcinoma, advancing prostate cancer usually becomes hormone-refractory. We attempted to establish a new prostate cancer therapy by controlling the malignancy of tumor cells through the induction of differentiation in vitro. METHODS We examined the ability of staurosporine to induce differentiation of human prostate cancer TSU-Pr1 cells into the cells with neuronal characteristics. RESULTS At low concentrations, staurosporine remarkably suppressed proliferation of human prostate cancer TSU-Pr1 cells without increasing dead cell number. TSU-Pr1 cells treated with 10(-8) M staurosporine began to extend neurites within 1 day, and approximately 80% of cells were changed to a neuronal morphology at 3 days. The expression of mRNA of tau, a microtubule-associated protein that is one of the essential components of neurite outgrowth, time-dependently increased in the cells treated with 10(-8) M staurosporine. Similarly, the amount of acetylcholinesterase increased. Colony-forming activity of TSU-Pr1 cells treated with 10(-8) M staurosporine for 7 days was 40% that of control cells. The invasive ability of TSU-Pr1 cells treated with staurosporine to penetrate through a reconstituted basement membrane of Matrigel was 20% that of untreated cells. CONCLUSIONS These results suggest that staurosporine might induce differentiation of human prostate cancer TSU-Pr1 cells to cells with neuronal characteristics.
Collapse
Affiliation(s)
- N Takahashi
- Department of Hygiene-Chemistry, Faculty of Pharmaceutical Sciences, Science University of Tokyo, Japan
| | | | | |
Collapse
|
166
|
van Leenders G, Dijkman H, Hulsbergen-van de Kaa C, Ruiter D, Schalken J. Demonstration of intermediate cells during human prostate epithelial differentiation in situ and in vitro using triple-staining confocal scanning microscopy. J Transl Med 2000; 80:1251-8. [PMID: 10950116 DOI: 10.1038/labinvest.3780133] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In human prostate epithelium, morphologically basal and luminal cells can be discriminated. The basal cell layer that putatively contains progenitor cells of the secretory epithelium is characterized by the expression of keratins (K) 5 and 14. Luminal cells represent the secretory compartment of the epithelium and express K8 and 18. We developed a technique for the simultaneous analysis of K5, 14, and 18 to identify intermediate cell stages in the prostate epithelium and to study the dynamic aspects of its differentiation in vitro. Nonmalignant prostate tissue and primary epithelial cultures were immunohistochemically characterized using triple staining with antibodies for K5, K14, and K18. Antibodies for K18 and K5 were conjugated directly with fluorochromes Alexa 488 and 546. K14 was visualized indirectly with streptavidin-Cy5. Keratin expression was analyzed by confocal scanning microscopy. The occurrence of exocrine and neuroendocrine differentiation in culture was determined via antibodies to prostate-specific antigen (PSA), chromogranin A, and serotonin. We found that basal cells expressed either K5(++)/14(++)/18+ or K5(++)/18+. The majority of luminal cells expressed K18(++), but colocalization of K5+/18(++) were recognized. Epithelial monolayer cultures predominantly revealed the basal cell phenotype K5(++)/14(++)/18+, whereas intermediate subpopulations expressing K5+/14+/18(++) and K5+/18(++) were also identified. On confluence, differentiation was induced as multicellular gland-like buds, and extensions became evident on top of the monolayer. These structures were composed of K18(++)- and K5+/18(+)-positive cell clusters surrounded by phenotypically basal cells. Few multicellular structures and cells in the monolayer showed exocrine differentiation (PSA+), but expression of chromogranin A and serotonin was absent. We conclude that simultaneous evaluation of keratin expression is useful for analyzing epithelial differentiation in the prostate. During this process, putative stem cells phenotypically resembling K5(++)/14(++)/18+ differentiate toward luminal cells (K18(++)) via intermediate cell stages, as identified by up-regulation of K18 and down-regulation of K5 and 14.
Collapse
Affiliation(s)
- G van Leenders
- Department of Pathology, University of Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
167
|
Schwede F, Maronde E, Genieser H, Jastorff B. Cyclic nucleotide analogs as biochemical tools and prospective drugs. Pharmacol Ther 2000; 87:199-226. [PMID: 11008001 DOI: 10.1016/s0163-7258(00)00051-6] [Citation(s) in RCA: 193] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cyclic AMP (cAMP) and cyclic GMP (cGMP) are key second messengers involved in a multitude of cellular events. From the wealth of synthetic analogs of cAMP and cGMP, only a few have been explored with regard to their therapeutic potential. Some of the first-generation cyclic nucleotide analogs were promising enough to be tested as drugs, for instance N(6),O(2)'-dibutyryl-cAMP and 8-chloro-cAMP (currently in clinical Phase II trials as an anticancer agent). Moreover, 8-bromo and dibutyryl analogs of cAMP and cGMP have become standard tools for investigations of biochemical and physiological signal transduction pathways. The discovery of the Rp-diastereomers of adenosine 3',5'-cyclic monophosphorothioate and guanosine 3',5'-cyclic monophosphorothioate as competitive inhibitors of cAMP- and cGMP-dependent protein kinases, as well as subsequent development of related analogs, has proven very useful for studying the molecular basis of signal transduction. These analogs exhibit a higher membrane permeability, increased resistance against degradation, and improved target specificity. Furthermore, better understanding of signaling pathways and ligand/protein interactions has led to new therapeutic strategies. For instance, Rp-8-bromo-adenosine 3',5'-cyclic monophosphorothioate is employed against diseases of the immune system. This review will focus mainly on recent developments in cyclic nucleotide-related biochemical and pharmacological research, but also highlights some historical findings in the field.
Collapse
Affiliation(s)
- F Schwede
- Center for Environmental Research and Environmental Technology, Department of Bioorganic Chemistry, University of Bremen, Leobener Strasse, D-28359, Bremen, Germany
| | | | | | | |
Collapse
|
168
|
Cox ME, Deeble PD, Bissonette EA, Parsons SJ. Activated 3',5'-cyclic AMP-dependent protein kinase is sufficient to induce neuroendocrine-like differentiation of the LNCaP prostate tumor cell line. J Biol Chem 2000; 275:13812-8. [PMID: 10788503 DOI: 10.1074/jbc.275.18.13812] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuroendocrine (NE) differentiation within prostate tumors is proposed to be a contributing factor in disease progression. However, the cellular origin and molecular mechanism controlling differentiation of prostatic NE cells are unresolved. The prostate tumor cell line, LNCaP, can reversibly acquire many NE characteristics in response to treatment with beta-adrenergic receptor agonists and activators of adenylate cyclase. In this study, we demonstrate that these treatments induce protein kinase A (PKA) activation in LNCaP cells and that ectopic expression of a constitutively activated form of the PKA catalytic subunit, CIalpha, results in acquisition of NE characteristics, including the extension of neuritic processes, cessation of mitotic activity, and production of neuron-specific enolase. Forskolin-, epinephrine-, and isoproterenol-dependent NE differentiation of LNCaP cells was significantly inhibited by expressing a dominant negative mutant of the PKA regulatory subunit, RIalpha. These results demonstrate that prostatic NE differentiation in response to these agents depends on PKA activation, and this signaling pathway may provide a therapeutic target for treating advanced forms of prostate cancer.
Collapse
Affiliation(s)
- M E Cox
- Department of Microbiology, University of Virginia School of Medicine, University of Virginia Health Systems, Charlottesville, Virginia 22908, USA.
| | | | | | | |
Collapse
|
169
|
Somlyo AV, Bradshaw D, Ramos S, Murphy C, Myers CE, Somlyo AP. Rho-kinase inhibitor retards migration and in vivo dissemination of human prostate cancer cells. Biochem Biophys Res Commun 2000; 269:652-9. [PMID: 10720471 DOI: 10.1006/bbrc.2000.2343] [Citation(s) in RCA: 182] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The Rho-kinase inhibitor, Y-27632, inhibited in vitro chemotactic migration to bone marrow fibroblast conditioned media and metastatic growth in immune-compromised mice of highly invasive human prostatic cancer (PC3) cells. Y-27632 also reduced myosin light chain phosphorylation and markedly altered the morphology of cells that developed numerous processes containing microtubules. A strikingly different, rounded phenotype was induced by an inhibitor of myosin light chain kinase, ML9. The M(110-130) subunit of the myosin phosphatase that is regulated by Rho-kinase was present in PC3 cells that contained significantly more RhoA than the less invasive, LNCaP cells. Y-27632 also inhibited angiogenesis as measured by endothelial cell tube formation on Matrigel. We conclude that invasiveness of human prostate cancer is facilitated by the Rho/Rho-kinase pathway, and exploration of selective Rho-kinase inhibitors for limiting invasive progress of prostate cancer is warranted.
Collapse
Affiliation(s)
- A V Somlyo
- Department of Molecular Physiology and Biological Physics, University of Virginia Health System, Charlottesville, Virginia 22906, USA.
| | | | | | | | | | | |
Collapse
|
170
|
Abstract
BACKGROUND In the human prostate cancer cell line LNCaP, interleukin (IL)-6 has been shown to regulate both growth and neuroendocrine (NE) differentiation. We recently observed that IL-6 mediated growth arrest in LNCaP by activating STAT 3. Since differentiation and growth arrest are often associated processes, we investigated whether STAT3 also mediated NE differentiation in this prostate cancer cell line. METHODS We treated previously characterized clones LNCaP-neo (neomycin-resistant LNCaP) and LNCaP-SF (LNCaP-STAT3 dominant negative mutant) with IL-6 and screened for NE differentiation by observing morphological changes and immunoblotting for two NE markers, neuron-specific enolase (NSE) and chromogranin A (ChA). To characterize further the role of STAT3 in growth arrest and differentiation, we transfected a wild-type STAT3 vector into PC-3 cells and generated a subclone PC-3-S3. In this clone, we assessed differentiation by observing morphological changes and determined growth responses by cell counting and clonogenic assays. RESULTS We observed that IL-6 induced formation of neurite extensions, morphologic features associated with NE differentiation, and enhanced expression of neuronal markers ChA and NSE in LNCaP-neo cells. In contrast, LNCaP-SF, possessing a dominant negative mutant form of STAT3, exhibited no characteristics of IL-6 induced NE differentiation. Furthermore, expression of a constitutively phosphorylated wild-type STAT3 in PC-3 cells inhibited growth and induced the formation of neurite extensions and NSE expression. CONCLUSIONS These results indicate that STAT3 is a mediator of both NE differentiation and growth inhibition in LNCaP and PC-3, suggesting a connection between growth inhibition and NE differentiation in prostate cancer.
Collapse
Affiliation(s)
- M T Spiotto
- Department of Radiation and Cellular Oncology and Division of Biological Sciences, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
171
|
TRANSDIFFERENTIATION OF PROSTATE CANCER CELLS TO A NEUROENDOCRINE CELL PHENOTYPE IN VITRO AND IN VIVO. J Urol 1999. [DOI: 10.1097/00005392-199911000-00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
172
|
BURCHARDT TATJANA, BURCHARDT MARTIN, CHEN MINWEI, CAO YICHEN, DE LA TAILLE ALEXANDRE, SHABSIGH AHMED, HAYEK OMAR, DORAI THAMBI, BUTTYAN RALPH. TRANSDIFFERENTIATION OF PROSTATE CANCER CELLS TO A NEUROENDOCRINE CELL PHENOTYPE IN VITRO AND IN VIVO. J Urol 1999. [DOI: 10.1016/s0022-5347(05)68241-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- TATJANA BURCHARDT
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| | - MARTIN BURCHARDT
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| | - MIN-WEI CHEN
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| | - YICHEN CAO
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| | - ALEXANDRE DE LA TAILLE
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| | - AHMED SHABSIGH
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| | - OMAR HAYEK
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| | - THAMBI DORAI
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| | - RALPH BUTTYAN
- From the Department of Urology and the Department of Pathology of the College of Physicians and Surgeons of Columbia University, New York, New York
| |
Collapse
|
173
|
Whitesell L, Liu X, Bagatell R. Inhibition of oncogene expression using vector-generated RNA antisense. Methods 1999; 18:296-303. [PMID: 10454988 DOI: 10.1006/meth.1999.0787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antisense RNA expression vectors have been developed relatively recently as a means to study the role of specific oncogenes in malignant transformation. In this paper, strategies for the construction of antisense plasmid vectors from commercially available reagents are described. Techniques for the introduction of these vectors into cell lines and tumors are also described and preferred methods for the evaluation of biological effects are presented. Lastly, using specific examples, the limitations and potential artifacts associated with antisense vector use in the study of tumorigenesis are discussed.
Collapse
Affiliation(s)
- L Whitesell
- Department of Pediatrics, University of Arizona, Tucson, Arizona 85724, USA.
| | | | | |
Collapse
|
174
|
Zi X, Agarwal R. Silibinin decreases prostate-specific antigen with cell growth inhibition via G1 arrest, leading to differentiation of prostate carcinoma cells: implications for prostate cancer intervention. Proc Natl Acad Sci U S A 1999; 96:7490-5. [PMID: 10377442 PMCID: PMC22113 DOI: 10.1073/pnas.96.13.7490] [Citation(s) in RCA: 165] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/1998] [Accepted: 04/26/1999] [Indexed: 11/18/2022] Open
Abstract
Reduction in serum prostate-specific antigen (PSA) levels has been proposed as an endpoint biomarker for hormone-refractory human prostate cancer intervention. We examined whether a flavonoid antioxidant silibinin (an active constituent of milk thistle) decreases PSA levels in hormone-refractory human prostate carcinoma LNCaP cells and whether this effect has biological relevance. Silibinin treatment of cells grown in serum resulted in a significant decrease in both intracellular and secreted forms of PSA concomitant with a highly significant to complete inhibition of cell growth via a G1 arrest in cell cycle progression. Treatment of cells grown in charcoal-stripped serum and 5alpha-dihydrotestosterone showed that the observed effects of silibinin are those involving androgen-stimulated PSA expression and cell growth. Silibinin-induced G1 arrest was associated with a marked decrease in the kinase activity of cyclin-dependent kinases (CDKs) and associated cyclins because of a highly significant decrease in cyclin D1, CDK4, and CDK6 levels and an induction of Cip1/p21 and Kip1/p27 followed by their increased binding with CDK2. Silibinin treatment of cells did not result in apoptosis and changes in p53 and bcl2, suggesting that the observed increase in Cip1/p21 is a p53-independent effect that does not lead to an apoptotic cell death pathway. Conversely, silibinin treatment resulted in a significant neuroendocrine differentiation of LNCaP cells as an alternative pathway after Cip1/p21 induction and G1 arrest. Together, these results suggest that silibinin could be a useful agent for the intervention of hormone-refractory human prostate cancer.
Collapse
Affiliation(s)
- X Zi
- Center for Cancer Causation and Prevention, AMC Cancer Research Center, 1600 Pierce Street, Denver, CO 80214, USA
| | | |
Collapse
|
175
|
Mitra SP, Carraway RE. Synergistic effects of neurotensin and beta-adrenergic agonist on 3,5-cyclic adenosine monophosphate accumulation and DNA synthesis in prostate cancer PC3 cells. Biochem Pharmacol 1999; 57:1391-7. [PMID: 10353260 DOI: 10.1016/s0006-2952(99)00064-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Since neurotensin is often co-stored with catecholamines and since it can excite the release of dopamine and norepinephrine, responses to this peptide might depend upon the activity of catecholaminergic systems. In this study, we used prostate cancer PC3 cells, which express neurotensin receptors and 12-adrenergic receptors, to demonstrate that neurotensin can potentiate the effects of isoproterenol on 3',5'-cyclic adenosine monophosphate (cAMP) formation and on inhibition of DNA synthesis. While neurotensin had only a slight effect on basal cAMP levels, it nearly doubled the response to isoproterenol even at maximal levels without altering potency. Neurotensin increased the rate of cAMP accumulation and the steady-state level achieved. Consistent with the known antimitogenic action of dibutyryl-cAMP in PC3 cells, isoproterenol was found to inhibit DNA synthesis concentration-dependently, measured using [3H]thymidine. Neurotensin enhanced DNA synthesis when given alone. However, it inhibited DNA synthesis when given with a threshold level of isoproterenol, which by itself had no significant effect. These results, demonstrating cross-talk in the neurotensin and beta-adrenergic signaling pathways, suggest that there may be other physiologic instances of similar interactions between neurotensin and catecholamines.
Collapse
Affiliation(s)
- S P Mitra
- Department of Physiology, University of Massachusetts Medical Center, Worcester 01655-0127, USA
| | | |
Collapse
|
176
|
Goto T, Matsushima H, Kasuya Y, Hosaka Y, Kitamura T, Kawabe K, Hida A, Ohta Y, Simizu T, Takeda K. The effect of papaverine on morphologic differentiation, proliferation and invasive potential of human prostatic cancer LNCaP cells. Int J Urol 1999; 6:314-9. [PMID: 10404308 DOI: 10.1046/j.1442-2042.1999.00069.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Intracellular cyclic adenosine monophosphate (AMP) level changes are thought to play an important role in inhibiting cell proliferation and inducing differentiation in several types of cells. It has been reported that cyclic AMP analogs induce terminal differentiation in human prostate cancer cells. Consequently, phosphodiesterase inhibitors may be useful in delineating the role of cyclic AMP in the differentiation of these cells. Therefore, the effect of phosphodiesterase inhibitors on morphologic differentiation, proliferation and invasive potential of human prostate cancer cells was investigated. METHODS Three human prostate cancer cell lines PC-3, DU145 and LNCaP were treated with one of the phosphodiesterase inhibitors, papaverine, 3-isobutyl-1-methylxanthine (IBMX) or theophylline, for 6 days. Morphologic changes of these cells induced by phosphodiesterase inhibitors were observed by microscopy. Intracellular cyclic AMP levels in LNCaP cells were measured by radioimmunoassay using a cyclic AMP assay kit. The effect of papaverine on the proliferation and invasive potential of LNCaP cells were measured by cell counting and the Matrigel invasion chamber assay. RESULTS Of the three agents, examined papaverine (10(-5) mol/L) is the most effective inducer of morphologic change and also raised intracellular cyclic AMP levels in LNCaP cells. However, unlike LNCaP cells, PC-3 and DU145 cells treated with phosphodiesterase inhibitors, including papaverine, showed little change in morphology. Additionally, proliferation and invasive potential of LNCaP cells were significantly inhibited by papaverine. CONCLUSION The results suggest that papaverine induces terminal differentiation in LNCaP cells, which is correlated with an intracellular cyclic AMP-mediated pathway.
Collapse
Affiliation(s)
- T Goto
- Faculty of Pharmaceutical Sciences, Science University of Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Abstract
BACKGROUND Information is presented on prostatic neuroendocrine cells and neuroendocrine differentiation in prostatic carcinoma. The prognostic and therapeutic implications of neuroendocrine differentiation in prostatic carcinoma are reviewed. METHODS Data are presented that support the intriguing link between neuroendocrine differentiation, tumor progression, and androgen-independent prostate cancer. The hormones, and the receptors, expressed by prostatic neuroendocrine cells are investigated in order to elucidate their significance for prognosis and therapy. RESULTS The prognostic significance of neuroendocrine differentiation in prostatic malignancy has been controversial, but recent studies employing markers such as chromogranin A and neuron-specific enolase suggest that neuroendocrine differentiation, as reflected by increased tissue expression and/or blood levels of these neuroendocrine secretory products, correlates with poor prognosis, tumor progression, and androgen-independence. Since all malignant neuroendocrine cells are devoid of androgen receptors and since neuroendocrine phenotypic expression is not suppressed by androgen ablation, clonal propagation of androgen receptor-negative neuroendocrine cells may play an important role in the pathway towards the androgen-independent state of prostatic carcinoma. This would have significant implications for the treatment of prostate cancer, as several of the hormones known to be expressed by neuroendocrine-differentiated, malignant prostatic cells are potential candidates for drug therapy. A limited number of hormones have been tested in this context, in particular somatostatin, bombesin, and serotonin. CONCLUSIONS Neuroendocrine differentiation in carcinoma of the prostate appears to be associated with poor prognosis, tumor progression, and the androgen-independent state, for which there is currently no successful therapy. Therefore, new therapeutic protocols and trials need to be developed to test drugs based on neuroendocrine hormones and/or their antagonists. An evaluation of this new therapeutic approach against prostatic carcinoma with neuroendocrine differentiation, including hormone-refractory cancer, is easily justified, since these tumors are unresponsive to current modes of therapy.
Collapse
Affiliation(s)
- P A Abrahamsson
- Department of Urology, University Hospitals of Malmö and Lund, University of Lund, Sweden.
| |
Collapse
|
178
|
Mori S, Murakami-Mori K, Bonavida B. Interleukin-6 induces G1 arrest through induction of p27(Kip1), a cyclin-dependent kinase inhibitor, and neuron-like morphology in LNCaP prostate tumor cells. Biochem Biophys Res Commun 1999; 257:609-14. [PMID: 10198259 DOI: 10.1006/bbrc.1999.0515] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prostate carcinoma cells express high levels of interleukin-6 (IL-6) and IL-6 receptor. In this study, we examined the effect of IL-6 on LNCaP human prostate carcinoma cells. IL-6 induces G1 growth arrest of LNCaP. Following IL-6 treatment of LNCaP, Western blot analysis showed that the protein levels of cyclin-dependent kinase-2 (CDK2), CDK4, and CDK6 were decreased, while accumulation of CDK inhibitor p27(Kip1) was rapidly and markedly induced. In vitro kinase assays revealed that the CDK-associated histone H1 and CDK4- and CDK6-associated pRb kinase activities were significantly inhibited in IL-6-treated LNCaP. Further, a significant amount of p27(Kip1) was co-precipitated with CDK2, CDK4 and CDK6, as detected in immunoprecipitation experiments. Thus, IL-6-induced G1 arrest appears to be due to the accumulation of p27(Kip1). In addition, IL-6-treated LNCaP cells induced neuron-like morphological changes. Since neuroendocrine differentiation is observed in most prostate carcinomas, these findings raise the possibility that IL-6 may be involved in neuroendocrine differentiation in vivo.
Collapse
Affiliation(s)
- S Mori
- Department of Microbiology and Immunology, UCLA School of Medicine, 10833 Le Conte Avenue, Los Angeles, California, 90095-1747, USA
| | | | | |
Collapse
|
179
|
Lee J, Choi YH, Nguyen P, Kim JS, Lee SJ, Trepel JB. Cyclic AMP induces inhibition of cyclin A expression and growth arrest in human hepatoma cells. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1449:261-8. [PMID: 10209305 DOI: 10.1016/s0167-4889(99)00019-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Classical cytotoxic therapy has been minimally useful in the treatment of hepatocellular carcinoma. In an effort to develop a new approach to the treatment of this neoplasm, we have investigated the signal transduction pathways regulating the growth of human hepatoma cells. In the data reported here, cyclic AMP (cAMP), a negative growth regulator for many cells of epithelial origin, induced G1 synchronization and apoptosis in the HepG2 human hepatoma cell line. The effects of cAMP on the components of the G1/S transition were analyzed. There was no detectable effect of two different cAMP analogs, 8-bromo cAMP or dibutyryl cAMP on the level of the D-type cyclins, cyclin E, cyclin-dependent kinase 2, cyclin-dependent kinase 4, p53, or the cyclin-dependent kinase inhibitors p21 or p27. In contrast, the cAMP analogs induced a dramatic downregulation of cyclin A protein, cyclin A messenger RNA, and cyclin A-dependent kinase activity. Cyclin A-dependent kinase has been shown to be required for the G1-S transition. Furthermore, cyclin A deregulation has been implicated in the pathogenesis of hepatocellular carcinoma. The data reported here suggest a novel signal transduction-based approach to hepatoma therapy.
Collapse
Affiliation(s)
- J Lee
- Medicine Branch, Division of Clinical Sciences, National Cancer Institute, National Institutes of Health, Building 10, Room 12N230, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
180
|
Jang JS, Lee SJ, Choi YH, Nguyen PM, Lee J, Hwang SG, Wu ML, Takano E, Maki M, Henkart PA, Trepel JB. Posttranslational regulation of the retinoblastoma gene family member p107 by calpain protease. Oncogene 1999; 18:1789-96. [PMID: 10086333 DOI: 10.1038/sj.onc.1202497] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The retinoblastoma protein plays a critical role in regulating the G1/S transition. Less is known about the function and regulation of the homologous pocket protein p107. Here we present evidence for the posttranslational regulation of p107 by the Ca2+-activated protease calpain. Three negative growth regulators, the HMG-CoA reductase inhibitor lovastatin, the antimetabolite 5-fluorouracil, and the cyclic nucleotide dibutyryl cAMP were found to induce cell type-specific loss of p107 protein which was reversible by the calpain inhibitor leucyl-leucyl-norleucinal but not by the serine protease inhibitor phenylmethylsulfonylfluoride, caspase inhibitors, or lactacystin, a specific inhibitor of the 26S proteasome. Purified calpain induced Ca2+-dependent p107 degradation in cell lysates. Transient expression of the specific calpain inhibitor calpastatin blocked the loss of p107 protein in lovastatin-treated cells, and the half-life of p107 was markedly lengthened in lovastatian-treated cells stably transfected with a calpastatin expression vector versus cells transfected with vector alone. The data presented here demonstrate down-regulation of p107 protein in response to various antiproliferative signals, and implicate calpain in p107 posttranslational regulation.
Collapse
Affiliation(s)
- J S Jang
- Medicine Branch, Division of Clinical Sciences, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Culig Z, Hobisch A, Herold M, Hittmair A, Thurnher M, Eder IE, Cronauer MV, Rieser C, Ramoner R, Bartsch G, Klocker H, Konwalinka G. Interleukin 1beta mediates the modulatory effects of monocytes on LNCaP human prostate cancer cells. Br J Cancer 1998; 78:1004-11. [PMID: 9792142 PMCID: PMC2063143 DOI: 10.1038/bjc.1998.619] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Proliferative and secretory responses in androgen-sensitive prostate cancer LNCaP cells are regulated by steroid and peptide hormones and by differentiation-promoting substances. In the present study, we evaluated whether peripheral blood monocytes that exhibit anti-tumour activity in haematopoietic and solid tumours influence growth and secretion in the LNCaP cell line. For this purpose, LNCaP cells were incubated with monocyte-conditioned medium (MCM), and proliferation as well as expression of androgen receptor (AR) and secretion of prostate-specific antigen (PSA) were assessed. Conditioned medium from monocytes reduced proliferation in a dose-dependent manner. Incubation with 40% MCM caused a 50% reduction in cell proliferation. AR protein decreased by 70% and PSA levels in supernatants from LNCaP cells were reduced by approximately 80% following treatment with MCM. We focused on the contribution of two major products of activated monocytes, prostaglandin E2 and interleukin 1beta (IL-1beta), to the MCM modulatory action. LNCaP cells treated with prostaglandin E2 showed neither a reduction in proliferation nor a down-regulation of AR and PSA levels. The effects of MCM on cellular proliferation, AR protein and PSA secretion were abolished by pretreatment of MCM with a neutralizing anti-IL-1beta antibody. In addition, recombinant IL-1beta was able to replace MCM for the inhibition of proliferation and down-regulation of AR and PSA proteins. LNCaP cells were shown to express the IL-1beta receptor type 1, which transduces IL-1beta signal. Our findings reveal that monocyte-derived IL-1beta inhibits the proliferation of androgen-responsive prostate tumour cells and reduces AR and PSA levels.
Collapse
Affiliation(s)
- Z Culig
- Department of Urology, University of Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Ueyama H, Ohkubo I. The expression of beta-microseminoprotein gene is regulated by cAMP. Biochem Biophys Res Commun 1998; 248:852-7. [PMID: 9704017 DOI: 10.1006/bbrc.1998.9064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The promoter of the human gene for beta-microseminoprotein (MSP) was analyzed by transient transfection assay using LNCaP cells. The region between -2738 and -207 was not essential for the basal promoter activity, but regions from -206, -108, and -83 were necessary. Forskolin induced promoter activity to 3- to 14-fold when constructions containing the possible cAMP response element (CRE, -60 to -53) were used, but did not do so when it was deleted from them. In a gel-mobility shift assay using a probe corresponding to -68 to -48, the shifted band was competed with a consensus CRE, and supershifted when the anti-CREB-1 antibody was included. Forskolin added to LNCaP cells increased MSP mRNA to 6-fold within 12 h. These results suggest that the expression of MSP gene is regulated by cAMP via the CRE present in the promoter. Hormones such as (-)-epinephrine or vasoactive intestinal polypeptide also increased MSP mRNA to 2- to 3-fold.
Collapse
Affiliation(s)
- H Ueyama
- Department of Medical Biochemistry, Shiga University of Medical Science, Otsu, Japan
| | | |
Collapse
|
183
|
Ekmekcioglu C, Feyertag J, Marktl W. Cinnamic acid inhibits proliferation and modulates brush border membrane enzyme activities in Caco-2 cells. Cancer Lett 1998; 128:137-44. [PMID: 9683274 DOI: 10.1016/s0304-3835(98)00073-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The objective of the present study was to examine whether cinnamic acid exerts antitumor activity against colon cancer cells in vitro. For this purpose we investigated the effect of cinnamic acid on cell proliferation and on the differentiation markers alkaline phosphatase, sucrase and aminopeptidase N in human colon adenocarcinoma cells (Caco-2). Cinnamic acid (2.5-8.0 mM) prolonged the doubling time and inhibited the DNA synthesis of growing cells. The antiproliferative effect occurred rapidly after 2 h of treatment with 8.0 mM cinnamic acid and reached nearly maximal values after 8 h of treatment. Sucrase and aminopeptidase N activities were stimulated under cinnamic acid treatment (4.0-8.0 mM), while alkaline phosphatase activity was inhibited in postconfluent cells (8.0 mM). Similar effects on enzyme activities were seen in non-proliferating cells. Cinnamic acid did not alter the adhesion to collagen matrix or cell viability. Intracellular cAMP levels were decreased significantly after 1 h of treatment with 8.0 mM cinnamic acid, suggesting that cinnamic acid induces its effects on enzyme activities partly by modulating the cAMP signaling pathway.
Collapse
Affiliation(s)
- C Ekmekcioglu
- Department of Medical Physiology, University of Vienna Medical School, Austria.
| | | | | |
Collapse
|
184
|
Griffiths K, Denis L, Turkes A, Morton MS. Possible relationship between dietary factors and pathogenesis of prostate cancer. Int J Urol 1998; 5:195-213. [PMID: 9624549 DOI: 10.1111/j.1442-2042.1998.tb00591.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- K Griffiths
- Tenovus Cancer Research Centre, University of Wales College of Medicine, Cardiff, UK
| | | | | | | |
Collapse
|
185
|
Lee SJ, Ha MJ, Lee J, Nguyen P, Choi YH, Pirnia F, Kang WK, Wang XF, Kim SJ, Trepel JB. Inhibition of the 3-hydroxy-3-methylglutaryl-coenzyme A reductase pathway induces p53-independent transcriptional regulation of p21(WAF1/CIP1) in human prostate carcinoma cells. J Biol Chem 1998; 273:10618-23. [PMID: 9553123 DOI: 10.1074/jbc.273.17.10618] [Citation(s) in RCA: 133] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Progression through the cell cycle is controlled by the induction of cyclins and the activation of cognate cyclin-dependent kinases. The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor lovastatin induces growth arrest and cell death in certain cancer cell types. We have pursued the mechanism of growth arrest in PC-3-M cells, a p53-null human prostate carcinoma cell line. Lovastatin treatment increased protein and mRNA levels of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1), increased binding of p21 with Cdk2, markedly inhibited cyclin E- and Cdk2-associated phosphorylation of histone H1 or GST-retinoblastoma protein, enhanced binding of the retinoblastoma protein to the transcription factor E2F-1 in vivo, and induced the activation of a p21 promoter reporter construct. By using p21 promoter deletion constructs, the lovastatin-responsive element was mapped to a region between -93 and -64 relative to the transcription start site. Promoter mutation analysis indicated that the lovastatin-responsive site coincided with the previously identified transforming growth factor-beta-responsive element. These data indicate that in human prostate carcinoma cells an inhibitor of the HMG-CoA reductase pathway can circumvent the loss of wild-type p53 function and induce critical downstream regulatory events leading to transcriptional activation of p21.
Collapse
Affiliation(s)
- S J Lee
- Medicine Branch, Division of Clinical Sciences, NCI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Qiu Y, Robinson D, Pretlow TG, Kung HJ. Etk/Bmx, a tyrosine kinase with a pleckstrin-homology domain, is an effector of phosphatidylinositol 3'-kinase and is involved in interleukin 6-induced neuroendocrine differentiation of prostate cancer cells. Proc Natl Acad Sci U S A 1998; 95:3644-9. [PMID: 9520419 PMCID: PMC19889 DOI: 10.1073/pnas.95.7.3644] [Citation(s) in RCA: 194] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/1997] [Accepted: 01/28/1998] [Indexed: 02/06/2023] Open
Abstract
Etk/Bmx is the newest member of Btk tyrosine kinase family that contains a pleckstrin homology domain, an src homology 3 domain, an src homology 2 domain, and a catalytic domain. Unlike other members of the Btk family kinases, which are mostly hemopoietic cell-specific, Etk/Bmx is preferentially expressed in epithelial and endothelial cells. We first identified this kinase in prostate cancer [Robinson, D., He, F., Pretlow, T. & Kung, H. J. (1996) Proc. Natl. Acad. Sci. USA 93, 5958-5962). Here we report that Etk is engaged in phosphatidylinositol 3-kinase (PI3-kinase) pathway and plays a pivotal role in interleukin 6 (IL-6) signaling in a prostate cancer cell line, LNCaP. Our evidence that PI3-kinase is involved in Etk activation includes: (i) Wortmannin, a specific inhibitor of PI3-kinase, abolished the activation of Etk by IL-6; (ii) a constitutively active p110 subunit of PI3-kinase was able to activate Etk in the absence of IL-6; and (iii) a dominant negative p85 subunit of PI3-kinase mutant blocked the activation of Etk by IL-6. Interestingly, IL-6 treatment of LNCaP induced a remarkable neuroendocrine-like differentiation phenotype, with neurite extension and enhanced expression of neuronal markers. This phenotype could be abrogated by the overexpression of a dominant-negative Etk, indicating Etk is required for this differentiation process.
Collapse
Affiliation(s)
- Y Qiu
- Department of Molecular Biology and Microbiology, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
187
|
Krijnen JL, Bogdanowicz JF, Seldenrijk CA, Mulder PG, van der Kwast TH. The prognostic value of neuroendocrine differentiation in adenocarcinoma of the prostate in relation to progression of disease after endocrine therapy. J Urol 1997; 158:171-4. [PMID: 9186347 DOI: 10.1097/00005392-199707000-00054] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE We evaluated the prognostic impact of neuroendocrine differentiation in prostate cancer with regard to the onset of endocrine therapy failure. MATERIALS AND METHODS A retrospective study was performed on 72 transurethral resection specimens from patients who subsequently underwent endocrine therapy for prostate cancer and were followed for 44 to 95 months. Progression-free interval was recorded. Distribution pattern and proportion of neuroendocrine cells were examined in transurethral resection specimens. Neuroendocrine cells were identified based on immunoreactivity for chromogranin A. RESULTS Of 32 patients with progressive disease 27 died of prostate cancer. Chromogranin A positive cells were found in 40 of the 72 prostate cancers (55%). In a Cox proportional hazards analysis neuroendocrine differentiation of the tumor showed a negative correlation with progression-free survival (p = 0.022), which proved to be independent of the Gleason score (p < 0.001). CONCLUSIONS Our results support the view that neuroendocrine differentiation in prostatic adenocarcinomas is a prognostic factor for progressive disease under subsequent endocrine therapy. This prognosticator acts independently of tumor grade.
Collapse
Affiliation(s)
- J L Krijnen
- Department of Pathology, Erasmus University, Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
188
|
Abstract
BACKGROUND Morphological, proliferative, and genetic changes were studied in androgen-responsive LNCaP cells in response to growth in charcoal-stripped (CS) media. METHODS AND RESULTS Within 5 days of treatment, there were dramatic changes in the morphology and organization of LNCaP cells. The cells unclumped and acquired a distinct neuronal-like appearance with small cell bodies and multiple long, thin processes. Despite this appearance, the cells stained negative to monoclonal antibodies to neuronal markers such as microtubule-associated protein-2 (MAP-2) and glial fibrillary acidic protein (GFAP). In situ end-labeling assay indicated that the number of cells showing signs of apoptosis (DNA fragmentation) increased dramatically in CS media compared to the control. However, ultrastructural changes and the fragmented DNA ladder that are used to define apoptosis were not observed. Instead of cell death, the cells became cytostatic, which can be reversed, although not completely, by exogeneous addition of dihydrotestosterone in a dose-dependent manner. Presence of mRNA of several genes involved in the apoptotic process, i.e., Bcl-2, Bcl-X, ICE, Ich-1, and DAD-1, was studied in response to normal and CS media. We detected mRNA of Bcl-2, Bcl-XL, Bcl-XS, Ich-1L and DAD-1, while ICE and Ich-1S were not expressed in LNCaP cells. CONCLUSIONS This suggests that certain signals that may be essential for complete execution of the apoptotic program may be missing in this in vitro model. This may explain our observation that the growth of LNCaP cells in CS media does not fully mimic castration-mediated regression of the prostate gland in vivo.
Collapse
Affiliation(s)
- B Saeed
- Pharmaceutical Products Division, Abbott Laboratories, Abbott Park, IL 60064, USA
| | | | | |
Collapse
|
189
|
Jiang L, Foster FM, Ward P, Tasevski V, Luttrell BM, Conigrave AD. Extracellular ATP triggers cyclic AMP-dependent differentiation of HL-60 cells. Biochem Biophys Res Commun 1997; 232:626-30. [PMID: 9126325 DOI: 10.1006/bbrc.1997.6345] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Extracellular ATP and ATP gamma S (1-1000 microM) stimulated cyclic AMP (cAMP) production in undifferentiated HL-60 cells. The potency order for adenine nucleotides and adenosine was ATP gamma S > ATP > > ADP > 3 AMP = Adenosine. Indomethacin (50 microM) had no effect on ATP-induced cAMP production. ATP and ATP gamma S also suppressed cell growth and induced differentiation as revealed by fMLP-stimulated beta-glucuronidase release 48 h after exposure. The potency order for the induction of fMLP-stimulated beta-glucuronidase release by adenine nucleotides and adenosine was ATP gamma S > 3 ATP > ADP > AMP = Adenosine approximately 0. The protein kinase A inhibitor Rp-8-Br-cAMPS (10-200 mM) suppressed ATP-induced differentiation but had no effect on ATP-dependent growth suppression. UTP which, like ATP, activates P2U receptors on HL-60 cells, had no effect on cAMP production, cell growth, or differentiation. The data suggest the existence of a novel receptor for ATP on undifferentiated HL-60 cells that is coupled to the activation of adenylate cyclase and cAMP-dependent differentiation.
Collapse
Affiliation(s)
- L Jiang
- Department of Biochemistry, University of Sydney, New South Wales, Australia
| | | | | | | | | | | |
Collapse
|
190
|
Noordzij MA, van Weerden WM, de Ridder CM, van der Kwast TH, Schröder FH, van Steenbrugge GJ. Neuroendocrine differentiation in human prostatic tumor models. THE AMERICAN JOURNAL OF PATHOLOGY 1996; 149:859-71. [PMID: 8780390 PMCID: PMC1865158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neuroendocrine (NE) cells can be identified in benign and malignant prostatic epithelia. Factors regulating their presence and their functions are poorly understood, mainly due to a lack of suitable experimental models. Fifteen in vitro and in vivo prostatic cancer tumor models, including a number of newly established in vivo models, were studied immunohistochemically for the presence of NE cells under different hormonal conditions. None of the in vitro models (PC-3, DU 145, LNCaP, and TSU) contained NE cells. Five of the seven xenograft models established at this laboratory contained NE cells. In three of these, NE cells were found only in the initial mouse passages. In the other two (PC-295 and PC-310), the NE phenotype was stable. NE features were confirmed by transmission electron microscopy and by Western analysis of chromogranin A expression. Immunohistochemical double-labeling experiments confirmed that NE cells in prostate cancer are post-mitotic (no Ki-67 expression) and do not express the androgen receptor. In the PC-295 and PC-310 models, short-term androgen withdrawal resulted in a rapidly increased number of NE cells. A time course experiment with PC-295-bearing mice strongly suggests that this increase occurred by induction of NE differentiation rather than by rapid proliferation and subsequent differentiation or selective persistence. In conclusion, these models are suitable to resolve fundamental questions with regard to the presence and functions of NE cells in human prostate cancer.
Collapse
Affiliation(s)
- M A Noordzij
- Department of Urology, Erasmus University, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
191
|
Harper ME, Glynne-Jones E, Goddard L, Thurston VJ, Griffiths K. Vascular endothelial growth factor (VEGF) expression in prostatic tumours and its relationship to neuroendocrine cells. Br J Cancer 1996; 74:910-6. [PMID: 8826857 PMCID: PMC2074752 DOI: 10.1038/bjc.1996.456] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) expression was examined by immunohistochemistry in 45 prostatic carcinoma specimens and ten benign prostatic tumours (BPH). The majority of carcinoma specimens exhibited cytoplasmic staining for VEGF and showed a trend of increasing expression with dedifferentiation (2p = 0.003). Immunoreactive VEGF was also seen in the prostatic carcinoma cell lines, the order of staining intensity was PC3 > DU145 > LNCaP. Intense granular cytoplasmic staining for VEGF was observed in neuroendocrine-like cells which were seen focally in many of the prostatic specimens. Consecutive sections were incubated with a chromogranin A antibody to confirm the neuroendocrine phenotype of these cells. A significant correlation (P < 0.0001) between the total number of intensely stained VEGF-positive cells and chromogranin A-positive cells was found. A subpopulation of neuroendocrine-like cells also showed intense immunoreactivity for transforming growth factor alpha (TGF-alpha). A correlation was observed (2p = 0.0092) between the intensity of VEGF and TGF-alpha immunostaining in carcinoma cells which were not of neuroendocrine differentiation. The presence of these two angiogenic factors may aid the neovascularisation of carcinomas and their increased expression in tumour-associated neuroendocrine cells may contribute to a more aggressive phenotype.
Collapse
Affiliation(s)
- M E Harper
- Tenovus Cancer Research Centre, University of Wales College of Medicine, UK
| | | | | | | | | |
Collapse
|
192
|
Liu AY, Corey E, Bladou F, Lange PH, Vessella RL. Prostatic cell lineage markers: emergence of BCL2+ cells of human prostate cancer xenograft LuCaP 23 following castration. Int J Cancer 1996; 65:85-9. [PMID: 8543402 DOI: 10.1002/(sici)1097-0215(19960103)65:1<85::aid-ijc15>3.0.co;2-h] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A model of prostate cancer progression based on the expression pattern of informative genes in the human prostate cancer xenograft LuCaP 23.1 is presented. Apparently, there are at least 2 tumor cell populations of LuCaP 23.1, representing 2 different phenotypes. One is NSE (neuron-specific enolase)-positive and the other NSE-negative. NSE-positive tumors were recovered after hormone-independent growth in castrated mice. These hormone-independent tumors also expressed BCL2, a gene product shown to inhibit apoptosis. With NSE, BCL2 and PSA (prostate-specific antigen) as identifying markers, the model specifies a putative progression sequence of the prostate cancer cell types. We also show a proposed lineage relationship among the 3 principal normal cell types found in the prostatic epithelium.
Collapse
Affiliation(s)
- A Y Liu
- Department of Urology, University of Washington, Seattle 98195, USA
| | | | | | | | | |
Collapse
|
193
|
|