51
|
Peixoto CA, Gomes FODS. The role of phosphodiesterase-5 inhibitors in prostatic inflammation: a review. JOURNAL OF INFLAMMATION-LONDON 2015; 12:54. [PMID: 26379476 PMCID: PMC4570643 DOI: 10.1186/s12950-015-0099-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 08/31/2015] [Indexed: 12/18/2022]
Abstract
Clinical and basic experimental evidence indicates that chronic inflammation is the greatest factor in benign prostatic hyperplasia (BPH) progression, which is the most common cause of Lower Urinary Tract Symptoms (LUTS). The use of anti-inflammatory agents such as steroids, cyclooxygenase-2 (COX-2) and phytotherapics have been investigated as forms of treatment for various prostate diseases. Recent evidence has demonstrated that PDE5 inhibitors (PDE5Is) improve symptoms of BPH/LUTS, possibly as a result of the relaxing of the smooth muscle fibers of the bladder and prostate by NO/cGMPc signaling, or by improving RhoA/Rho-kinase (ROCK), and reduction of the hyperactivity of the autonomic nervous system. However, some results have suggested that besides vasodilatation and their anti-proliferative effect, PDE5Is exert a direct anti-inflammatory effect, by raising cGMP. Given that inflammation is major factor in benign prostatic hyperplasia (BPH) progression, PDE5Is could act also restore prostatic function as they act as potent anti-inflammatory drugs. This review aims to provide a comprehensive summary of the use of phosphodiesterase-5 inhibitors to treat prostatic inflammation.
Collapse
Affiliation(s)
- Christina Alves Peixoto
- Laboratório de Ultraestrutura, Centro de Pesquisas Aggeu Magalhães (CPqAM-FIOCRUZ), Fundação Oswaldo Cruz, Av. Moraes Rego s/n, CEP: 50670-420, Cidade Universitária, Recife, PE Brazil
| | - Fabiana Oliveira Dos Santos Gomes
- Laboratório de Ultraestrutura, Centro de Pesquisas Aggeu Magalhães (CPqAM-FIOCRUZ), Fundação Oswaldo Cruz, Av. Moraes Rego s/n, CEP: 50670-420, Cidade Universitária, Recife, PE Brazil
| |
Collapse
|
52
|
Hu CD, Choo R, Huang J. Neuroendocrine differentiation in prostate cancer: a mechanism of radioresistance and treatment failure. Front Oncol 2015; 5:90. [PMID: 25927031 PMCID: PMC4396194 DOI: 10.3389/fonc.2015.00090] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/26/2015] [Indexed: 12/20/2022] Open
Abstract
Neuroendocrine differentiation (NED) in prostate cancer is a well-recognized phenotypic change by which prostate cancer cells transdifferentiate into neuroendocrine-like (NE-like) cells. NE-like cells lack the expression of androgen receptor and prostate specific antigen, and are resistant to treatments. In addition, NE-like cells secrete peptide hormones and growth factors to support the growth of surrounding tumor cells in a paracrine manner. Accumulated evidence has suggested that NED is associated with disease progression and poor prognosis. The importance of NED in prostate cancer progression and therapeutic response is further supported by the fact that therapeutic agents, including androgen-deprivation therapy, chemotherapeutic agents, and radiotherapy, also induce NED. We will review the work supporting the overall hypothesis that therapy-induced NED is a mechanism of resistance to treatments, as well as discuss the relationship between therapy-induced NED and therapy-induced senescence, epithelial-to-mesenchymal transition, and cancer stem cells. Furthermore, we will use radiation-induced NED as a model to explore several NED-based targeting strategies for development of novel therapeutics. Finally, we propose future studies that will specifically address therapy-induced NED in the hope that a better treatment regimen for prostate cancer can be developed.
Collapse
Affiliation(s)
- Chang-Deng Hu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University Center for Cancer Research, Purdue University , West Lafayette, IN , USA
| | - Richard Choo
- Department of Radiation Oncology, Mayo Clinic , Rochester, MN , USA
| | - Jiaoti Huang
- Department of Pathology, David Geffen School of Medicine at UCLA , Los Angeles, CA , USA
| |
Collapse
|
53
|
Grigore AD, Ben-Jacob E, Farach-Carson MC. Prostate cancer and neuroendocrine differentiation: more neuronal, less endocrine? Front Oncol 2015; 5:37. [PMID: 25785244 PMCID: PMC4347593 DOI: 10.3389/fonc.2015.00037] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/03/2015] [Indexed: 12/17/2022] Open
Abstract
Neuroendocrine differentiation (NED) marks a structural and functional feature of certain cancers, including prostate cancer (PCa), whereby the malignant tissue contains a significant proportion of cells displaying neuronal, endocrine, or mixed features. NED cells produce, and can secrete, a cocktail of mediators commonly encountered in the nervous system, which may stimulate and coordinate cancer growth. In PCa, NED appears during advanced stages, subsequent to treatment, and accompanies treatment resistance and poor prognosis. However, the term “neuroendocrine” in this context is intrinsically vague. This article seeks to provide a framework on which a unified view of NED might emerge. First, we review the mutually beneficial interplay between PCa and neural structures, mainly supported by cell biology experiments and neurological conditions. Next, we address the correlations between PCa and neural functions, as described in the literature. Based upon the integration of clinical and basic observations, we suggest that it is legitimate to seek for true neural differentiation, or neuromimicry, in cancer progression, most notably in PCa cells exhibiting what is commonly described as NED.
Collapse
Affiliation(s)
- Alexandru Dan Grigore
- Department of BioSciences, Rice University , Houston, TX , USA ; Center for Theoretical Biological Physics, Rice University , Houston, TX , USA
| | - Eshel Ben-Jacob
- Center for Theoretical Biological Physics, Rice University , Houston, TX , USA ; Sackler School of Physics and Astronomy, Tel Aviv University , Tel Aviv , Israel ; Sagol School of Neuroscience, Tel Aviv University , Tel Aviv , Israel
| | - Mary C Farach-Carson
- Department of BioSciences, Rice University , Houston, TX , USA ; Center for Theoretical Biological Physics, Rice University , Houston, TX , USA ; Department of Bioengineering, Rice University , Houston, TX , USA
| |
Collapse
|
54
|
Expression and functional role of orphan receptor GPR158 in prostate cancer growth and progression. PLoS One 2015; 10:e0117758. [PMID: 25693195 PMCID: PMC4333349 DOI: 10.1371/journal.pone.0117758] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/23/2014] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second-leading cause of cancer-related mortality, after lung cancer, in men from developed countries. In its early stages, primary tumor growth is dependent on androgens, thus generally can be controlled by androgen deprivation therapy (ADT). Eventually however, the disease progresses to castration-resistant prostate cancer (CRPC), a lethal form in need of more effective treatments. G-protein coupled receptors (GPCRs) comprise a large clan of cell surface proteins that have been implicated as therapeutic targets in PCa growth and progression. The findings reported here provide intriguing evidence of a role for the newly characterized glutamate family member GPR158 in PCa growth and progression. We found that GPR158 promotes PCa cell proliferation independent of androgen receptor (AR) functionality and that this requires its localization in the nucleus of the cell. This suggests that GPR158 acts by mechanisms different from other GPCRs. GPR158 expression is stimulated by androgens and GPR158 stimulates AR expression, implying a potential to sensitize tumors to low androgen conditions during ADT via a positive feedback loop. Further, we found GPR158 expression correlates with a neuroendocrine (NE) differentiation phenotype and promotes anchorage-independent colony formation implying a role for GPR158 in therapeutic progression and tumor formation. GPR158 expression was increased at the invading front of prostate tumors that formed in the genetically defined conditional Pten knockout mouse model, and co-localized with elevated AR expression in the cell nucleus. Kaplan-Meier analysis on a dataset from the Memorial Sloan Kettering cancer genome portal showed that increased GPR158 expression in tumors is associated with lower disease-free survival. Our findings strongly suggest that pharmaceuticals targeting GPR158 activities could represent a novel and innovative approach to the prevention and management of CRPC.
Collapse
|
55
|
Weaver EM, Zamora FJ, Puplampu-Dove YA, Kiessu E, Hearne JL, Martin-Caraballo M. Regulation of T-type calcium channel expression by sodium butyrate in prostate cancer cells. Eur J Pharmacol 2015; 749:20-31. [DOI: 10.1016/j.ejphar.2014.12.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/15/2014] [Accepted: 12/15/2014] [Indexed: 10/24/2022]
|
56
|
Braadland PR, Ramberg H, Grytli HH, Taskén KA. β-Adrenergic Receptor Signaling in Prostate Cancer. Front Oncol 2015; 4:375. [PMID: 25629002 PMCID: PMC4290544 DOI: 10.3389/fonc.2014.00375] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/16/2014] [Indexed: 12/29/2022] Open
Abstract
Enhanced sympathetic signaling, often associated with obesity and chronic stress, is increasingly acknowledged as a contributor to cancer aggressiveness. In prostate cancer, intact sympathetic nerves are critical for tumor formation, and sympathectomy induces apoptosis and blocks tumor growth. Perineural invasion, involving enrichment of intra-prostatic nerves, is frequently observed in prostate cancer and is associated with poor prognosis. β2-adrenergic receptor (ADRB2), the most abundant receptor for sympathetic signals in prostate luminal cells, has been shown to regulate trans-differentiation of cancer cells to neuroendocrine-like cells and to affect apoptosis, angiogenesis, epithelial–mesenchymal transition, migration, and metastasis. Epidemiologic studies have shown that use of β-blockers, inhibiting β-adrenergic receptor activity, is associated with reduced prostate cancer-specific mortality. In this review, we aim to present an overview on how β-adrenergic receptor and its downstream signaling cascade influence the development of aggressive prostate cancer, primarily through regulating neuroendocrine differentiation.
Collapse
Affiliation(s)
- Peder Rustøen Braadland
- Department of Tumor Biology, Institute of Cancer Research, Division of Cancer Medicine, Transplantation and Surgery, Oslo University Hospital , Oslo , Norway
| | - Håkon Ramberg
- Department of Tumor Biology, Institute of Cancer Research, Division of Cancer Medicine, Transplantation and Surgery, Oslo University Hospital , Oslo , Norway
| | - Helene Hartvedt Grytli
- Department of Tumor Biology, Institute of Cancer Research, Division of Cancer Medicine, Transplantation and Surgery, Oslo University Hospital , Oslo , Norway
| | - Kristin Austlid Taskén
- Department of Tumor Biology, Institute of Cancer Research, Division of Cancer Medicine, Transplantation and Surgery, Oslo University Hospital , Oslo , Norway ; Institute of Clinical Medicine, University of Oslo , Oslo , Norway
| |
Collapse
|
57
|
Courel M, El Yamani FZ, Alexandre D, El Fatemi H, Delestre C, Montero-Hadjadje M, Tazi F, Amarti A, Magoul R, Chartrel N, Anouar Y. Secretogranin II is overexpressed in advanced prostate cancer and promotes the neuroendocrine differentiation of prostate cancer cells. Eur J Cancer 2014; 50:3039-49. [PMID: 25307750 DOI: 10.1016/j.ejca.2014.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/11/2014] [Accepted: 09/15/2014] [Indexed: 11/28/2022]
Abstract
AIM In prostate cancer (PCa), neuroendocrine differentiation (NED) is commonly observed in relapsing, hormone therapy-resistant tumours after androgen deprivation. However, the molecular mechanisms involved in the NED of PCa cells remain poorly understood. In this study, we investigated the expression of the neuroendocrine secretory protein secretogranin II (SgII) in PCa, and its potential involvement in the progression of this cancer as a granulogenic factor promoting NED. METHODS We have examined SgII immunoreactivity in 25 benign prostate hyperplasia and 32 PCa biopsies. In vitro experiments were performed to investigate the involvement of SgII in the neuroendocrine differentiation and the proliferation of PCa cell lines. RESULTS We showed that immunoreactive SgII intensity correlates with tumour grade in PCa patients. Using the androgen-dependent lymph node cancer prostate cells (LNCaP) cells, we found that NED triggered by androgen deprivation is associated with the induction of SgII expression. In addition, forced expression of SgII in LNCaP cells implemented a regulated secretory pathway by triggering the formation of secretory granule-like structures competent for hormone storage and regulated release. Finally, we found that SgII promotes prostate cancer (CaP) cell proliferation. CONCLUSION The present data show that SgII is highly expressed in advanced PCa and may contribute to the neuroendocrine differentiation by promoting the formation of secretory granules and the proliferation of PCa cells.
Collapse
Affiliation(s)
- Maïté Courel
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Fatima-Zohra El Yamani
- Laboratory of Neuroendocrinology and Nutritional and Climatic Environment, Faculty of Sciences Dhar-El Mahraz, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - David Alexandre
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Hinde El Fatemi
- Department of Pathology, University Hospital Hassan II, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - Charlène Delestre
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Maité Montero-Hadjadje
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Fadl Tazi
- Department of Pathology, University Hospital Hassan II, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - Afaf Amarti
- Department of Pathology, University Hospital Hassan II, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - Rabia Magoul
- Laboratory of Neuroendocrinology and Nutritional and Climatic Environment, Faculty of Sciences Dhar-El Mahraz, University Sidi Mohamed Ben Abdellah, 30000 Fez, Morocco
| | - Nicolas Chartrel
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France
| | - Youssef Anouar
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France; Normandy University, Normandy, France.
| |
Collapse
|
58
|
Tien JCY, Liao L, Liu Y, Liu Z, Lee DK, Wang F, Xu J. The steroid receptor coactivator-3 is required for developing neuroendocrine tumor in the mouse prostate. Int J Biol Sci 2014; 10:1116-27. [PMID: 25332686 PMCID: PMC4202028 DOI: 10.7150/ijbs.10236] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/30/2014] [Indexed: 11/25/2022] Open
Abstract
Neuroendocrine tumor cells (NETCs) are commonly observed in prostate cancer. Their presence is associated with castration resistance, metastasis and poor prognosis. Cellular and molecular mechanisms for NETC initiation and growth are unknown. TRAMP mice develop heterogeneous adenocarcinomas induced by expression of the SV40-T/t oncogene in prostate epithelial cells. Here, we demonstrate prostate tumors in TRAMP mice with a mixed genetic background are characterized mostly by atypical hyperplasia (AH) containing steroid receptor coactiator-3-positive, androgen receptor-positive and synaptophysin-negative (SRC-3+/AR+/Syp-) cells. Few SRC-3+/AR-/Syp+ NETCs are present in their prostates. We generated TRAMP mice in which SRC-3 was specifically ablated in AR+/Syp- prostatic epithelial cells (termed PE3KOT mice). In these animals, we observed a substantial reduction in SRC-3-/AR+/Syp- AH tumor growth. There was a corresponding increase in SRC-3-/AR+/Syp- phyllodes lesions, suggesting SRC-3 knockout can convert aggressive AH tumors with mostly epithelial tumor cells into less aggressive phyllodes lesions with mostly stromal tissue. Surprisingly, PE3KOT mice developed many more SRC-3+/AR-/Syp+ NETCs versus control TRAMP mice, indicating SRC-3 expression was retained in NETCs. In contrast, TRAMP mice with global SRC-3 knockout did not develop any NETC, indicating SRC-3 is required for developing NETC. Analysis of cell-differentiating markers revealed that these NETCs might not be derived from the mature AR-/Syp+ neuroendocrine cells or the AR+/Syp- luminal epithelial tumor cells. Instead, these NETCs might originate from the SV40-T/t-transformed intermediate/progenitor epithelial cells. In summary, SRC-3 is required for both AR+/Syp- AH tumor growth and AR-/Syp+ NETC development, suggesting SRC-3 is a target for inhibiting aggressive prostate cancer containing NETCs.
Collapse
Affiliation(s)
- Jean Ching-Yi Tien
- 1. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA; ; 2. Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| | - Lan Liao
- 1. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yonghong Liu
- 1. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA; ; 2. Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| | - Zhaoliang Liu
- 1. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA; ; 2. Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| | - Dong-Kee Lee
- 1. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Fen Wang
- 2. Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| | - Jianming Xu
- 1. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA; ; 2. Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas 77030, USA; ; 3. Insitute for Cancer Medicine, Luzhou Medical College, Luzhou, Sichuan 646000, China
| |
Collapse
|
59
|
Figliozzi RW, Chen F, Balish M, Ajavon A, Hsia SV. Thyroid hormone-dependent epigenetic suppression of herpes simplex virus-1 gene expression and viral replication in differentiated neuroendocrine cells. J Neurol Sci 2014; 346:164-73. [PMID: 25175854 DOI: 10.1016/j.jns.2014.08.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/05/2014] [Accepted: 08/13/2014] [Indexed: 12/21/2022]
Abstract
A global HSV-1 gene repression occurs during latency in sensory neurons where most viral gene transcriptions are suppressed. The molecular mechanisms of gene silencing and how stress factors trigger the reactivation are not well understood. Thyroid hormones are known to be altered due to stress, and with its nuclear receptor impart transcriptional repression or activation depending upon the hormone level. Therefore we hypothesized that triiodothyronine (T3) treatment of infected differentiated neuron like cells would reduce the ability of HSV-1 to produce viral progeny compared to untreated infected cells. Previously we identified putative thyroid hormone receptor elements (TREs) within the promoter regions of HSV-1 thymidine kinase (TK) and other key genes. Searching for a human cell line that can model neuronal HSV-1 infection, we performed HSV-1 infection experiments on differentiated human neuroendocrine cells, LNCaP. Upon androgen deprivation these cells undergo complete differentiation and exhibit neuronal-like morphology and physiology. These cells were readily infected by our HSV-1 recombinant virus, expressing GFP and maintaining many processes iconic of dendritic morphology. Our results demonstrated that differentiated LNCaP cells produced suppressive effects on HSV-1 gene expression and replication compared to its undifferentiated counterpart and T3 treatment has further decreased the viral plaque counts compared to untreated cells. Upon washout of the T3 viral plaque counts were restored, indicating an increase of viral replication. The qRT-PCR experiments using primers for TK showed reduced expression under T3 treatment. ChIP assays using a panel of antibodies for H3 lysine 9 epigenetic marks showed increased repressive marks on the promoter regions of TK. In conclusion we have demonstrated a T3 mediated quiescent infection in differentiated LNCaP cells that has potential to mimic latent infection. In this HSV-1 infection model thyroid hormone treatment caused decreased viral replication, repressed TK expression and increased repressive histone tail marks on the TK promoter.
Collapse
Affiliation(s)
- Robert W Figliozzi
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, College of Pharmacy, Princess Anne, MD 21853, USA
| | - Feng Chen
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, College of Pharmacy, Princess Anne, MD 21853, USA
| | - Matthew Balish
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, College of Pharmacy, Princess Anne, MD 21853, USA
| | - Amakoe Ajavon
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, College of Pharmacy, Princess Anne, MD 21853, USA
| | - S Victor Hsia
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, College of Pharmacy, Princess Anne, MD 21853, USA.
| |
Collapse
|
60
|
Gomes FODS, Carvalho MDC, Saraiva KLA, Ribeiro EL, E Silva AKS, Donato MAM, Rocha SWS, Santos e Silva B, Peixoto CA. Effect of chronic Sildenafil treatment on the prostate of C57Bl/6 mice. Tissue Cell 2014; 46:439-49. [PMID: 25239757 DOI: 10.1016/j.tice.2014.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 06/23/2014] [Accepted: 08/01/2014] [Indexed: 10/24/2022]
Abstract
Sildenafil is a potent and selective inhibitor of phosphodiesterase-5 (PDE5) and is considered first-line therapy for erectile dysfunction. Nowadays, Sildenafil is used extensively throughout the world on patients with pulmonary hypertension. However, few studies have evaluated the possible side effects of chronic Sildenafil treatment on the male reproductive system, specifically in the prostate. In the present study, it was demonstrated via morphological and ultrastructural analysis that chronic treatment with Sildenafil induced an enhancement of the glandular activity of the prostate. In addition, mice treated with Sildenafil showed a significant increase in testosterone serum levels. However, no statistically significant differences were observed in nitric oxide serum levels, or in sGC, eNOS, PSA and TGF-β prostatic expression. In conclusion, the present study suggests that chronic use of Sildenafil does not cause evident prostatic damage, and therefore, can be used pharmacologically to treat a variety of disorders.
Collapse
Affiliation(s)
| | - Maria da Conceição Carvalho
- Laboratório de Microscopia e Microanálise do Centro de Tecnologias Estratégicas do Nordeste (CETENE), Brazil
| | | | - Edlene Lima Ribeiro
- Laboratório de Ultraestrutura do Instituto Aggeu Magalhães (FIOCRUZ), Brazil; Universidade Federal de Pernambuco (UFPE), Brazil
| | - Amanda Karolina Soares E Silva
- Laboratório de Ultraestrutura do Instituto Aggeu Magalhães (FIOCRUZ), Brazil; Universidade Federal de Pernambuco (UFPE), Brazil
| | - Mariana Aragão Matos Donato
- Laboratório de Ultraestrutura do Instituto Aggeu Magalhães (FIOCRUZ), Brazil; Universidade Federal de Pernambuco (UFPE), Brazil
| | - Sura Wanessa Santos Rocha
- Laboratório de Ultraestrutura do Instituto Aggeu Magalhães (FIOCRUZ), Brazil; Universidade Federal de Pernambuco (UFPE), Brazil
| | - Bruna Santos e Silva
- Laboratório de Ultraestrutura do Instituto Aggeu Magalhães (FIOCRUZ), Brazil; Universidade Federal de Pernambuco (UFPE), Brazil
| | | |
Collapse
|
61
|
Pernicová Z, Slabáková E, Fedr R, Šimečková Š, Jaroš J, Suchánková T, Bouchal J, Kharaishvili G, Král M, Kozubík A, Souček K. The role of high cell density in the promotion of neuroendocrine transdifferentiation of prostate cancer cells. Mol Cancer 2014; 13:113. [PMID: 24884804 PMCID: PMC4229954 DOI: 10.1186/1476-4598-13-113] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 05/13/2014] [Indexed: 11/13/2022] Open
Abstract
Background Tumor heterogeneity and the plasticity of cancer cells present challenges for effective clinical diagnosis and therapy. Such challenges are epitomized by neuroendocrine transdifferentiation (NED) and the emergence of neuroendocrine-like cancer cells in prostate tumors. This phenomenon frequently arises from androgen-depleted prostate adenocarcinoma and is associated with the development of castration-resistant prostate cancer and poor prognosis. Results In this study, we showed that NED was evoked in both androgen receptor (AR)-positive and AR-negative prostate epithelial cell lines by growing the cells to a high density. Androgen depletion and high-density cultivation were both associated with cell cycle arrest and deregulated expression of several cell cycle regulators, such as p27Kip1, members of the cyclin D protein family, and Cdk2. Dual inhibition of Cdk1 and Cdk2 using pharmacological inhibitor or RNAi led to modulation of the cell cycle and promotion of NED. We further demonstrated that the cyclic adenosine 3′, 5′-monophosphate (cAMP)-mediated pathway is activated in the high-density conditions. Importantly, inhibition of cAMP signaling using a specific inhibitor of adenylate cyclase, MDL-12330A, abolished the promotion of NED by high cell density. Conclusions Taken together, our results imply a new relationship between cell cycle attenuation and promotion of NED and suggest high cell density as a trigger for cAMP signaling that can mediate reversible NED in prostate cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, v,v,i, Královopolská 135, CZ-612 65 Brno, Czech Republic.
| |
Collapse
|
62
|
Terry S, Beltran H. The many faces of neuroendocrine differentiation in prostate cancer progression. Front Oncol 2014; 4:60. [PMID: 24724054 PMCID: PMC3971158 DOI: 10.3389/fonc.2014.00060] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 03/12/2014] [Indexed: 12/15/2022] Open
Abstract
In normal prostate, neuroendocrine (NE) cells are rare and interspersed among the epithelium. These cells are believed to provide trophic signals to epithelial cell populations through the secretion of an abundance of neuropeptides that can diffuse to influence surrounding cells. In the setting of prostate cancer (PC), NE cells can also stimulate surrounding prostate adenocarcinoma cell growth, but in some cases adenocarcinoma cells themselves acquire NE characteristics. This epithelial plasticity is associated with decreased androgen receptor (AR) signaling and the accumulation of neuronal and stem cell characteristics. Transformation to an NE phenotype is one proposed mechanism of resistance to contemporary AR-targeted treatments, is associated with poor prognosis, and thought to represent up to 25% of lethal PCs. Importantly, the advent of high-throughput technologies has started to provide clues for understanding the complex molecular profiles of tumors exhibiting NE differentiation. Here, we discuss these recent advances, the multifaceted manner by which an NE-like state may arise during the different stages of disease progression, and the potential benefit of this knowledge for the management of patients with advanced PC.
Collapse
Affiliation(s)
- Stéphane Terry
- U955, Institut Mondor de Recherche Biomédicale, INSERM , Créteil , France ; UMR 3244, Institut Curie , Paris , France
| | - Himisha Beltran
- Division of Hematology and Medical Oncology, Weill Cornell Medical College , New York, NY , USA
| |
Collapse
|
63
|
De Petrocellis L, Ligresti A, Schiano Moriello A, Iappelli M, Verde R, Stott CG, Cristino L, Orlando P, Di Marzo V. Non-THC cannabinoids inhibit prostate carcinoma growth in vitro and in vivo: pro-apoptotic effects and underlying mechanisms. Br J Pharmacol 2014; 168:79-102. [PMID: 22594963 DOI: 10.1111/j.1476-5381.2012.02027.x] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Cannabinoid receptor activation induces prostate carcinoma cell (PCC) apoptosis, but cannabinoids other than Δ(9) -tetrahydrocannabinol (THC), which lack potency at cannabinoid receptors, have not been investigated. Some of these compounds antagonize transient receptor potential melastatin type-8 (TRPM8) channels, the expression of which is necessary for androgen receptor (AR)-dependent PCC survival. EXPERIMENTAL APPROACH We tested pure cannabinoids and extracts from Cannabis strains enriched in particular cannabinoids (BDS), on AR-positive (LNCaP and 22RV1) and -negative (DU-145 and PC-3) cells, by evaluating cell viability (MTT test), cell cycle arrest and apoptosis induction, by FACS scans, caspase 3/7 assays, DNA fragmentation and TUNEL, and size of xenograft tumours induced by LNCaP and DU-145 cells. KEY RESULTS Cannabidiol (CBD) significantly inhibited cell viability. Other compounds became effective in cells deprived of serum for 24 h. Several BDS were more potent than the pure compounds in the presence of serum. CBD-BDS (i.p.) potentiated the effects of bicalutamide and docetaxel against LNCaP and DU-145 xenograft tumours and, given alone, reduced LNCaP xenograft size. CBD (1-10 µM) induced apoptosis and induced markers of intrinsic apoptotic pathways (PUMA and CHOP expression and intracellular Ca(2+)). In LNCaP cells, the pro-apoptotic effect of CBD was only partly due to TRPM8 antagonism and was accompanied by down-regulation of AR, p53 activation and elevation of reactive oxygen species. LNCaP cells differentiated to androgen-insensitive neuroendocrine-like cells were more sensitive to CBD-induced apoptosis. CONCLUSIONS AND IMPLICATIONS These data support the clinical testing of CBD against prostate carcinoma.
Collapse
Affiliation(s)
- Luciano De Petrocellis
- Istituto di Cibernetica, Endocannabinoid Research Group, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Harada T, Giorgio L, Harris TJ, Pham DT, Ngo HT, Need EF, Coventry BJ, Lincoln SF, Easton CJ, Buchanan G, Kee TW. Diamide Linked γ-Cyclodextrin Dimers as Molecular-Scale Delivery Systems for the Medicinal Pigment Curcumin to Prostate Cancer Cells. Mol Pharm 2013; 10:4481-90. [DOI: 10.1021/mp400309s] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Takaaki Harada
- Department
of Chemistry, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Lauren Giorgio
- Basil
Hetzel Institute for Translational Health Research, Discipline of
Surgery, Queen Elizabeth Hospital, The University of Adelaide, Woodville South, South Australia, 5011, Australia
| | - Tiffany J. Harris
- Basil
Hetzel Institute for Translational Health Research, Discipline of
Surgery, Queen Elizabeth Hospital, The University of Adelaide, Woodville South, South Australia, 5011, Australia
| | - Duc-Truc Pham
- Department
of Chemistry, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Huy Tien Ngo
- Department
of Chemistry, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Eleanor F. Need
- Basil
Hetzel Institute for Translational Health Research, Discipline of
Surgery, Queen Elizabeth Hospital, The University of Adelaide, Woodville South, South Australia, 5011, Australia
| | - Brendon J. Coventry
- Discipline
of Surgery, Royal Adelaide Hospital, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Stephen F. Lincoln
- Department
of Chemistry, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Christopher J. Easton
- Research
School of Chemistry, Australian National University, Canberra, Australian Capital Territory, 0200, Australia
| | - Grant Buchanan
- Basil
Hetzel Institute for Translational Health Research, Discipline of
Surgery, Queen Elizabeth Hospital, The University of Adelaide, Woodville South, South Australia, 5011, Australia
| | - Tak W. Kee
- Department
of Chemistry, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| |
Collapse
|
65
|
Rapa I, Volante M, Migliore C, Farsetti A, Berruti A, Vittorio Scagliotti G, Giordano S, Papotti M. Human ASH-1 promotes neuroendocrine differentiation in androgen deprivation conditions and interferes with androgen responsiveness in prostate cancer cells. Prostate 2013; 73:1241-9. [PMID: 23657976 DOI: 10.1002/pros.22679] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 04/03/2013] [Indexed: 01/26/2023]
Abstract
BACKGROUND Neuroendocrine differentiation in prostate cancer is a dynamic process associated to the onset of hormone-refractory disease in vivo. The molecular mechanisms underlying this process are poorly recognized. Our study aimed at testing in vitro the role of hASH-1, a transcription factor implicated in neuroendocrine differentiation, in the onset of neuroendocrine phenotype in prostate cancer cells. METHODS Androgen sensitive LNCAP, androgen insensitive PC-3, and three immortalized prostate cancer cell lines were cultured in standard and androgen deprivation conditions. Expression of hASH-1 was modulated by either specific lentiviral transduction or shRNA interference. Inhibitors of WNT-11, a WNT family member associated to the development of neuroendocrine differentiation in prostate cancer, were also used. Cell viability was measured using the MTS method. Neuroendocrine phenotype was assessed by morphology, immunohistochemistry and real time PCR for several neuroendocrine markers. RESULTS hASH-1 was up-modulated by androgen deprivation in LNCaP cells and in androgen-sensitive immortalized prostate cancer cells, and associated with the onset of a neuroendocrine phenotype. Silencing of hASH-1 prevented neuroendocrine differentiation, as did also the selective interference with the WNT-11 pathway. Moreover, hASH-1 over-expression in LNCaP cells was sufficient to promote neuroendocrine differentiation and increased cell viability at basal and androgen-deprived growth conditions. CONCLUSION In summary, the present data support previous evidence that the acquisition of a neuroendocrine phenotype is linked to androgen responsiveness profiles and suggest a pivotal role of hASH-1 transcription factor, whose activity might be explored as a potential therapeutic target in prostate cancer, with special reference to hormone refractory disease.
Collapse
Affiliation(s)
- Ida Rapa
- Department of Oncology at San Luigi Hospital, University of Turin, Orbassano, Turin, Italy
| | | | | | | | | | | | | | | |
Collapse
|
66
|
The molecular basis for ethnic variation and histological subtype differences in prostate cancer. SCIENCE CHINA-LIFE SCIENCES 2013; 56:780-7. [PMID: 23852643 PMCID: PMC4078990 DOI: 10.1007/s11427-013-4522-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/01/2013] [Indexed: 01/04/2023]
Abstract
Prostate cancer is a common malignancy among men in Western countries. Recently the morbidity and mortality of prostate cancer increase dramatically in several oriental countries including China. Rapidly evolving technology in molecular biology such as high-throughput sequencing and integrative analysis of genomic and transcriptomic landscapes have enabled the identification of key oncogenic events for prostate cancer initiation, progression and resistance to hormonal therapy. These surging data of prostate cancer genome also provide insights on ethnic variation and the differences in histological subtype of this disease. In this review, differences in the incidence of prostate cancer and the prevalence of main genetic alterations between Asian and Western populations are discussed. We also review the recent findings on the mechanisms underlying neuroendocrine differentiation of prostate cancer and the development of small cell neuroendocrine carcinoma after androgen deprivation therapy.
Collapse
|
67
|
Lin DC, Xu L, Ding LW, Sharma A, Liu LZ, Yang H, Tan P, Vadgama J, Karlan BY, Lester J, Urban N, Schummer M, Doan N, Said JW, Sun H, Walsh M, Thomas CJ, Patel P, Yin D, Chan D, Koeffler HP. Genomic and functional characterizations of phosphodiesterase subtype 4D in human cancers. Proc Natl Acad Sci U S A 2013; 110:6109-6114. [PMID: 23536305 PMCID: PMC3625360 DOI: 10.1073/pnas.1218206110] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Discovery of cancer genes through interrogation of genomic dosage is one of the major approaches in cancer research. In this study, we report that phosphodiesterase subtype 4D (PDE4D) gene was homozygously deleted in 198 cases of 5,569 primary solid tumors (3.56%), with most being internal microdeletions. Unexpectedly, the microdeletions did not result in loss of their gene products. Screening PDE4D expression in 11 different types of primary tumor samples (n = 165) with immunohistochemistry staining revealed that its protein levels were up-regulated compared with corresponding nontransformed tissues. Importantly, depletion of endogenous PDE4D with three independent shRNAs caused apoptosis and growth inhibition in multiple types of cancer cells, including breast, lung, ovary, endometrium, gastric, and melanoma, which could be rescued by reexpression of PDE4D. We further showed that antitumor events triggered by PDE4D suppression were lineage-dependently associated with Bcl-2 interacting mediator of cell death (BIM) induction and microphthalmia-associated transcription factor (MITF) down-regulation. Furthermore, ectopic expression of the PDE4D short isoform, PDE4D2, enhanced the proliferation of cancer cells both in vitro and in vivo. Moreover, treatment of cancer cells with a unique specific PDE4D inhibitor, 26B, triggered massive cell death and growth retardation. Notably, these antineoplastic effects induced by either shRNAs or small molecule occurred preferentially in cancer cells but not in nonmalignant epithelial cells. These results suggest that although targeted by genomic homozygous microdeletions, PDE4D functions as a tumor-promoting factor and represents a unique targetable enzyme of cancer cells.
Collapse
Affiliation(s)
- De-Chen Lin
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, University of California School of Medicine, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Vanoverberghe K, Lehen’kyi V, Thébault S, Raphaël M, Vanden Abeele F, Slomianny C, Mariot P, Prevarskaya N. Cytoskeleton reorganization as an alternative mechanism of store-operated calcium entry control in neuroendocrine-differentiated cells. PLoS One 2012; 7:e45615. [PMID: 23049826 PMCID: PMC3458093 DOI: 10.1371/journal.pone.0045615] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/23/2012] [Indexed: 02/07/2023] Open
Abstract
Neuroendocrine differentiation (NED) is a hallmark of advanced androgen-independent prostate cancer, for which no successful therapy exists. NED tumour cells escape apoptotic cell death by alterations of Ca(2+) homeostasis where the store-operated Ca(2+) entry (SOCE) is known to be a key event. We have previously shown that the downregulation of Orai1 protein representing the major molecular component of endogenous SOCE in human prostate cancer cells, and constituting the principal source of Ca(2+) influx used by the cell to trigger apoptosis, contributes to the establishment of an apoptosis-resistant phenotype (Cell Death Dis. 2010 Sep 16;1:e75.). Here, we report for the first time that the decrease of SOCE during NED may be caused by alternative NED-induced mechanism involving cytoskeleton reorganisation. NED induced by androgen deprivation resulted in a decrease of SOCE due to cortical F-actin over-polymerization which inhibits thapsigargin-induced SOCE. The disruption of F-actin polymerization by Cytochalasin D in NED cells restored SOCE, while the induction of F-actin polymerization by jasplakinolide or calyculin A diminished SOCE without changing the expression of key SOCE players: Orai1, STIM1, and TRPC1. Our data suggest that targeting cytoskeleton-induced pathways of malignant cells together with SOCE-involved channels may prove a useful strategy in the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Karine Vanoverberghe
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - V’yacheslav Lehen’kyi
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Stéphanie Thébault
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Maylis Raphaël
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Fabien Vanden Abeele
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Christian Slomianny
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Pascal Mariot
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Natalia Prevarskaya
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
- * E-mail:
| |
Collapse
|
69
|
Ge D, Gao AC, Zhang Q, Liu S, Xue Y, You Z. LNCaP prostate cancer cells with autocrine interleukin-6 expression are resistant to IL-6-induced neuroendocrine differentiation due to increased expression of suppressors of cytokine signaling. Prostate 2012; 72:1306-16. [PMID: 22213096 PMCID: PMC3665156 DOI: 10.1002/pros.22479] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 12/02/2011] [Indexed: 01/17/2023]
Abstract
BACKGROUND Neuroendocrine differentiation (NED) is one of the mechanisms underlying development of castration-resistant prostate cancer (CRPC). In this study, we investigated IL-6-induced NED in two LNCaP sublines. METHODS LNCaP-S17, an LNCaP subline that secretes IL-6, and LNCaP-C3, a control subline that does not express IL-6, were analyzed for IL-6-induced NED, activation of JAK2 and STAT3 pathways, and expression of IL-6/IL-6R signaling proteins and downstream target genes. RESULTS IL-6 did not induce NED in LNCaP-S17 cells, even though IL-6 induced NED in LNCaP-C3 cells. IL-6 activated JAK2 and STAT3 pathways in LNCaP-C3 cells but not in LNCaP-S17 cells. IL-6 did not activate ERK1/2, AKT, or NF-κB pathways in either cell line. Both LNCaP-C3 and LNCaP-S17 cell lines expressed IL-6R, gp130, and TYK2 at almost the same levels and did not express JAK1 or JAK3. The basal level of JAK2 expression was slightly higher in LNCaP-C3 cells than in LNCaP-S17 cells. Two suppressors of cytokine signaling, SOCS7 and cytokine-inducible SH2 protein (CIS), were expressed constitutively at higher levels in LNCaP-S17 cells than in LNCaP-C3 cells, while SOCS1 to SOCS6 were expressed at approximately the same levels. Using siRNA to knockdown SOCS7 and CIS expression in LNCaP-S17 cells led to increased phosphorylation of STAT3 upon IL-6 stimulation. CONCLUSIONS LNCaP-S17 cells are resistant to exogenous IL-6-induced NED due to increased levels of CIS/SOCS7 that block activation of JAK2-STAT3 pathways.
Collapse
Affiliation(s)
- Dongxia Ge
- Department of Structural & Cellular Biology, Department of Orthopaedic Surgery, Tulane Cancer Center, Louisiana Cancer Research Consortium, Tulane Center for Aging, Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Allen C. Gao
- Department of Urology, University of California at Davis Medical Center, Sacramento, California 95817
| | - Qiuyang Zhang
- Department of Structural & Cellular Biology, Department of Orthopaedic Surgery, Tulane Cancer Center, Louisiana Cancer Research Consortium, Tulane Center for Aging, Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Sen Liu
- Department of Structural & Cellular Biology, Department of Orthopaedic Surgery, Tulane Cancer Center, Louisiana Cancer Research Consortium, Tulane Center for Aging, Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Yun Xue
- Department of Structural & Cellular Biology, Department of Orthopaedic Surgery, Tulane Cancer Center, Louisiana Cancer Research Consortium, Tulane Center for Aging, Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Zongbing You
- Department of Structural & Cellular Biology, Department of Orthopaedic Surgery, Tulane Cancer Center, Louisiana Cancer Research Consortium, Tulane Center for Aging, Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112
- Correspondence to: Zongbing You, MD, PhD, Department of Structural & Cellular Biology, Tulane University School of Medicine, 1430 Tulane Ave SL 49, New Orleans, LA 70112. Fax: 504-988-1687; Tel: 504-988-0467;
| |
Collapse
|
70
|
Katori Y, Shibata S, Kawase T, Cho BH, Murakami G. Transient Appearance of Tyrosine Hydroxylase Immunoreactive Cells in the Midline Epithelial Seam of the Human Fetal Secondary Palate. Cleft Palate Craniofac J 2012; 49:414-24. [DOI: 10.1597/10-121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Objective Transient immunoreactivity for tyrosine hydroxylase, which mediates the conversion of the amino acid L-tyrosine to dihydroxyphenylalanine, in the midline epithelial seam between the bilateral palatal shelves was investigated in human fetuses. Materials and Methods Horizontal or frontal paraffin sections of two human fetuses at 9 and 15 weeks of gestation were used to examine the distribution of tyrosine hydroxylase–immunoreactive cells in regions of the entire head other than the brain. Immunohistochemical staining for S100 protein, calretinin, cytokeratin 14, and vimentin was examined using adjacent or near sections. Results Tyrosine hydroxylase–immunoreactive cells were large and densely distributed in the midline epithelial seam at the site of palatal fusion in fetuses at 9 weeks but not in fetuses at 15 weeks, in which the midline epithelial seam had already disappeared. No expression of S100 protein, calretinin, or vimentin was detected, but the midline epithelial seam was positive for cytokeratin 14. Tyrosine hydroxylase immunoreactivity was not detected in epithelia during the process of palatal fusion in mice from E 14.0 to 15.0. Conclusions These findings indicate that tyrosine hydroxylase–immunoreactive cells in the midline epithelial seams are nonneural epithelial cells and suggest that the tyrosine hydroxylase is a novel factor involved in normal palatal formation, especially the fate of the midline epithelial seam in humans.
Collapse
Affiliation(s)
| | - Shunichi Shibata
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Tetsuaki Kawase
- Department of Otorhinolaryngology, Laboratory of Rehabilitative Auditory Science, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Baik Hwan Cho
- Department of Surgery, Faculty of Medicine, Chonbuk National University, Jeonju, Jeonbuk, South Korea
| | - Gen Murakami
- Division of Internal Medicine, Iwamizawa Koujin-kai Hospital, Iwamizawa, Hokkaido, Japan
| |
Collapse
|
71
|
Abstract
Prostate cancer is initially androgen-dependent but, over time, usually develops hormone- and chemo-resistance. The present study investigated a role for p21-activated kinase 4 (PAK4) in prostate cancer progression. PAK4 activation was markedly inhibited by H89, a specific protein kinase A (PKA) inhibitor, and PAK4 was activated by the elevation of cAMP. The catalytic subunit of PKA interacted with the regulatory domain of PAK4, and directly phosphorylated PAK4 at serine 474 (S474). Catalytically active PAK4 enhanced the transcriptional activity of CREB independent of S133 phosphorylation. Stable knockdown of PAK4 in PC-3 and DU145 prostate cancer cells inhibited tumor formation in nude mice. Decreased tumorigenicity correlated with decreased expression of CREB and its targets, including Bcl-2 and cyclin A1. Additionally, in androgen-dependent LNCap-FGC cells, PAK4 regulated cAMP-induced neuroendocrine differentiation, which is known to promote tumor progression. Finally, PAK4 enhanced survival and decreased apoptosis following chemotherapy. These results suggested that PAK4 regulates progression toward hormone- and chemo-resistance in prostate cancer, and this study identified both a novel activation mechanism and potential downstream effector pathways. Therefore, PAK4 may be a promising therapeutic target in prostate cancer.
Collapse
|
72
|
Kashiwagi E, Shiota M, Yokomizo A, Itsumi M, Inokuchi J, Uchiumi T, Naito S. Downregulation of phosphodiesterase 4B (PDE4B) activates protein kinase A and contributes to the progression of prostate cancer. Prostate 2012; 72:741-51. [PMID: 22529021 DOI: 10.1002/pros.21478] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 07/29/2011] [Indexed: 11/05/2022]
Abstract
BACKGROUND Prostate cancer is the most commonly diagnosed non-cutaneous cancer in American men. Unfortunately, few successful therapies for castration-resistant prostate cancer (CRPC) exist. The protein kinase A (PKA) pathway is a critical mediator of cellular proliferation and differentiation in various normal and cancerous cells. However, the PKA activity and the mechanism of regulation in CRPC remain unclear. Then, in this study, we intended to reveal the PKA activity and the mechanism of regulation in CRPC. METHODS Western blotting, quantitative real-time polymerase chain reaction, cytotoxicity analysis, and cell proliferation assay were used to resolve the regulatory role of PKA in prostate cancer cell line, LNCaP and their derivatives. RESULTS cAMP-specific phosphodiesterase 4B (PDE4B) was downregulated and the PKA pathway was activated in castration-resistant LNCaP derivatives (CxR cells). Rolipram activated the PKA pathway via inhibition of PDE4B, resulting in AR transactivation while the PKA inhibitor, H89 reduced AR transactivation. In response to hydrogen peroxide and in hydrogen peroxide-resistant LNCaP derivatives (HPR50 cells) PDE4B was decreased and as a result PKA activity was increased. Moreover, PDE4B expression was reduced in advanced prostate cancer and PDE4B knockdown promoted castration-resistant growth of LNCaP cells. CONCLUSIONS Oxidative stress may suppress PDE4B expression and activate the PKA pathway. The PDE4B/PKA pathway contributed to progression of androgen-dependent prostate cancer to CRPC. This pathway may represent an attractive therapeutic molecular target.
Collapse
Affiliation(s)
- Eiji Kashiwagi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
73
|
Jones SE, Palmer TM. Protein kinase A-mediated phosphorylation of RhoA on serine 188 triggers the rapid induction of a neuroendocrine-like phenotype in prostate cancer epithelial cells. Cell Signal 2012; 24:1504-14. [PMID: 22504159 PMCID: PMC3510439 DOI: 10.1016/j.cellsig.2012.03.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 03/12/2012] [Accepted: 03/22/2012] [Indexed: 01/27/2023]
Abstract
Whilst androgen ablation therapy is used to treat locally advanced or metastatic forms of prostate cancer, side-effects can include the emergence of an androgen-independent neuroendocrine cell population which is associated with poor prognosis. Here we have examined how cyclic AMP elevation regulates early events in the neuroendocrine differentiation process. We demonstrate that selective activation of protein kinase A is necessary and sufficient for cyclic AMP (cAMP) elevation to rapidly promote a neuroendocrine phenotype in LNCaP cells independent of de novo protein synthesis. Furthermore, the effects of cAMP could be recapitulated by inhibition of RhoA signalling or pharmacological inhibition of Rho kinase. Conversely, expression of constitutively active Gln63Leu-mutated RhoA acted as a dominant-negative inhibitor of cAMP-mediated NE phenotype formation. Consistent with these observations, cAMP elevation triggered the PKA-dependent phosphorylation of RhoA on serine 188, and a non-phosphorylatable Ser188Ala RhoA mutant functioned as a dominant-negative inhibitor of cAMP-mediated neuroendocrine phenotype formation. These results suggest that PKA-mediated inhibition of RhoA via its phosphorylation on serine 188 and the subsequent inhibition of ROCK activity plays a key role in determining initial changes in cellular morphology during LNCaP cell differentiation to a neuroendocrine phenotype. It also raises the possibility that targeted suppression of this pathway to inhibit neuroendocrine cell expansion might be a useful adjuvant to conventional prostate cancer therapy.
Collapse
Affiliation(s)
- Sarah E Jones
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK.
| | | |
Collapse
|
74
|
Deng X, Elzey BD, Poulson JM, Morrison WB, Ko SC, Hahn NM, Ratliff TL, Hu CD. Ionizing radiation induces neuroendocrine differentiation of prostate cancer cells in vitro, in vivo and in prostate cancer patients. Am J Cancer Res 2011; 1:834-844. [PMID: 22016831 PMCID: PMC3196282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Accepted: 07/27/2011] [Indexed: 05/31/2023] Open
Abstract
Prostate cancer remains the most common noncutaneous cancer among American men. Although most patients can be cured by surgery and radiotherapy, 32,050 patients still died of the disease in 2010. Many patients receive radiotherapy either as a primary therapy, salvage therapy, or in combination with surgery or hormonal therapy. Despite initial treatment, several studies suggest that approximately 10% of low-risk prostate cancer patients and up to 30-60% with more advanced cancer patients experience biochemical recurrence within five years after radiotherapy. Thus, elucidating the molecular mechanisms underlying radioresistance and tumor recurrence has the potential to significantly reduce prostate cancer mortality. We previously demonstrated that fractionated ionizing radiation (IR) can induce the prostate cancer cell line LNCaP to undergo neuroendocrine differentiation (NED) by activation of cAMP response element binding protein (CREB) and cytoplasmic sequestration of ATF2, two CRE-binding transcription factors that oppose each other to regulate NED. Importantly, IR-induced NED is reversible and de-differentiated cells are cross-resistant to IR, androgen depletion and docetaxel treatments. These findings suggest that radiation-induced NED may allow prostate cancer cells to survive treatment and contribute to tumor recurrence. In the present study, we further demonstrated that IR also induces NED in a subset of DU-145 and PC-3 cells. In addition, we confirmed that IR induces NED in LNCaP xenograft tumors in nude mice, and observed that the plasma chro-mogranin A (CgA) level, a biomarker for NED, is increased by 2- to 5-fold in tumor-bearing mice after fractionated radiation doses of 20 and 40 Gy, respectively. Consistent with these in vivo findings, a pilot study in prostate cancer patients showed that the serum CgA level is elevated in 4 out of 9 patients after radiotherapy. Taken together, these findings provide evidence that radiation-induced NED is a general therapeutic response in a subset of prostate cancer patients. Thus, a large scale analysis of radiotherapy-induced NED in prostate cancer patients and its correlation to clinical outcomes will likely provide new insight into the role of NED in prostate cancer radiotherapy and prognosis.
Collapse
Affiliation(s)
- Xuehong Deng
- Department of Medicinal Chemistry and molecular pharmacology, Purdue UniversityWest Lafayette, IN 47907, USA
| | - Bennett D Elzey
- Department of Comparative Pathobiology, Purdue UniversityWest Lafayette, IN 47907, USA
| | - Jean M Poulson
- Department of Veterinary Clinical Sciences, Purdue UniversityWest Lafayette, IN 47907, USA
| | - Wallace B Morrison
- Department of Veterinary Clinical Sciences, Purdue UniversityWest Lafayette, IN 47907, USA
| | - Song-Chu Ko
- Department of Radiation Oncology, Indiana University Melvin and Bren Simon Cancer CenterIndianapolis, IN 46202, USA
| | - Noah M Hahn
- Department of Medicine, Indiana University Melvin and Bren Simon Cancer CenterIndianapolis, IN 46202, USA
| | - Timothy L Ratliff
- Department of Comparative Pathobiology, Purdue UniversityWest Lafayette, IN 47907, USA
| | - Chang-Deng Hu
- Department of Medicinal Chemistry and molecular pharmacology, Purdue UniversityWest Lafayette, IN 47907, USA
| |
Collapse
|
75
|
Merkle D, Hoffmann R. Roles of cAMP and cAMP-dependent protein kinase in the progression of prostate cancer: cross-talk with the androgen receptor. Cell Signal 2010; 23:507-15. [PMID: 20813184 DOI: 10.1016/j.cellsig.2010.08.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2010] [Revised: 08/14/2010] [Accepted: 08/20/2010] [Indexed: 01/22/2023]
Abstract
Prostate carcinomas are among the most frequently diagnosed and death causing cancers affecting males in the developed world. It has become clear that the molecular mechanisms that drive the differentiation of normal prostate cells towards neoplasia involve multiple signal transduction cascades that often overlap and interact. A critical mediator of cellular proliferation and differentiation in various cells (and cancers) is the cAMP-dependent protein kinase, also known as protein kinase A (PKA), and its activating secondary messenger, cAMP. PKA and cAMP have been shown to play critical roles in prostate carcinogenesis and are the subject of this review. In particular we will focus on the cross-talk between PKA/cAMP signaling and that of the androgen receptor.
Collapse
Affiliation(s)
- Dennis Merkle
- Philips Research, High Tech Campus, 5656AE, Eindhoven,The Netherlands.
| | | |
Collapse
|
76
|
Xiao X, Li BX, Mitton B, Ikeda A, Sakamoto KM. Targeting CREB for cancer therapy: friend or foe. Curr Cancer Drug Targets 2010; 10:384-91. [PMID: 20370681 DOI: 10.2174/156800910791208535] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2009] [Accepted: 03/31/2010] [Indexed: 11/22/2022]
Abstract
The cyclic-AMP response element-binding protein (CREB) is a nuclear transcription factor activated by phosphorylation at Ser133 by multiple serine/threonine (Ser/Thr) kinases. Upon phosphorylation, CREB binds the transcriptional co-activator, CBP (CREB-binding protein), to initiate CREB-dependent gene transcription. CREB is a critical regulator of cell differentiation, proliferation and survival in the nervous system. Recent studies have shown that CREB is involved tumor initiation, progression and metastasis, supporting its role as a proto-oncogene. Overexpression and over-activation of CREB were observed in cancer tissues from patients with prostate cancer, breast cancer, non-small-cell lung cancer and acute leukemia while down-regulation of CREB in several distinct cancer cell lines resulted in inhibition of cell proliferation and induction of apoptosis, suggesting that CREB may be a promising target for cancer therapy. Although CREB, as a transcription factor, is a challenging target for small molecules, various small molecules have been discovered to inhibit CREB phosphorylation, CREB-DNA, or CREB-CBP interaction. These results suggest that CREB is a suitable transcription factor for drug targeting and therefore targeting CREB could represent a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Xiangshu Xiao
- Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA.
| | | | | | | | | |
Collapse
|
77
|
Li B, Wang XY, Tian Z, Xiao XJ, Xu Q, Wei CX, Y F, Sun HC, Chen GH. Directional differentiation of chicken spermatogonial stem cells in vitro. Cytotherapy 2010; 12:326-31. [PMID: 20078389 DOI: 10.3109/14653240903518155] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Mammalian spermatogonial stem cells (SSC) are able to differentiate into different cell types in vitro, which are valuable sources for regenerative medicine and gene transfer studies. We investigated the differentiation potential of chicken SSC into osteoblasts, neuron-like cells and adipocytes in vitro. METHODS Chicken SSC from the testes of 18- and 20-day-old chicken embryos were cultured in different induction media for three passages in vitro. For differentiation into osteoblasts, SSC were cultured in Dulbecco's modified Eagle medium (DMEM) supplemented with 1 x 10(-4) micromol/mL desamethasone, 10 micromol/mL (beta-sodium glycerophosphate and 0.05 mg/mL vitamin C, and examined by microscopy after Von Kossa's, cytochemical and immunohistochemical staining. For differentiation into neuron-like cells, SSC were cultured in DMEM supplemented with 1 x 10(-3) micromol/mL retinoic acid (RA), 5.0 micromol/mL 3-isobutyl-1-methylxanthine (IBMX) and examined by microscopy after toluidine blue or immunohistochemical staining. For differentiation into adipocytes, SSC were cultured in DMEM supplemented with 1 x 10(-3) micromol/mL dexamethasone, 0.01 mg/mL insulin, 0.5 micromol/mL IBMX and examined by microscopy after Oil red O staining and reverse transcriptase-polymerase chain reaction (RT-PCR) for gene expression of peroxisome proliferation activation receptor-gamma (PPAR-gamma). RESULTS After 15 and 21 days of culture in the induction medium for osteoblast differentiation, 75% and 80% chicken SSC differentiated into osteoblasts, as confirmed by Von Kossa's, calcium-cobalt and collagen I antibody staining. After 3 and 7 days of culture in the induction medium for neuron-like cell differentiation, 78% and 85% SSC became neuron-like cells, as confirmed by staining with toluidine blue and the monoclonal antibody against neuron-specific enolase, nestin and glial fibrillary acidic protein. After 7 days of culture in the induction for adipocyte differentiation, 85% SSC differentiated into adipocytes, as confirmed by Oil red O staining and RT-PCT for PPAR-gamma gene expression. DISCUSSION Our results show that chicken SSC can differentiate into osteoblasts, neuron-like cells and adipocytes under similar conditions as for directional differentiation of mammalian SSC in vitro. The findings show the feasibility of using SSC-derived cells for developmental biology and gene transfer studies in chickens.
Collapse
Affiliation(s)
- Bichun Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Roles of db-cAMP, IBMX and RA in aspects of neural differentiation of cord blood derived mesenchymal-like stem cells. PLoS One 2010; 5:e9398. [PMID: 20195526 PMCID: PMC2827567 DOI: 10.1371/journal.pone.0009398] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 02/04/2010] [Indexed: 02/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have multilineage differentiation potential which includes cell lineages of the central nervous system; hence MSCs might be useful in the treatment of neurodegenerative diseases such as Parkinson's disease. Although mesenchymal stem cells have been shown to differentiate into the neural lineage, there is still little knowledge about the underlying mechanisms of differentiation particularly towards specialized neurons such as dopaminergic neurons. Here, we show that MSCs derived from human umbilical cord blood (MSChUCBs) are capable of expressing tyrosine hydroxylase (TH) and Nurr1, markers typically associated with DA neurons. We also found differential phosphorylation of TH isoforms indicating the presence of post-translational mechanisms possibly activating and modifying TH in MSChUCB. Furthermore, functional dissection of components in the differentiation medium revealed that dibutyryl-cAMP (db-cAMP), 3-isobutyl-1-methylxanthine (IBMX) and retinoic acid (RA) are involved in the regulation of Nurr1 and Neurofilament-L expression as well as in the differential phosphorylation of TH. We also demonstrate a possible inhibitory role of the protein kinase A signaling pathway in the phosphorylation of specific TH isoforms.
Collapse
|
79
|
Dayon A, Brizuela L, Martin C, Mazerolles C, Pirot N, Doumerc N, Nogueira L, Golzio M, Teissié J, Serre G, Rischmann P, Malavaud B, Cuvillier O. Sphingosine kinase-1 is central to androgen-regulated prostate cancer growth and survival. PLoS One 2009; 4:e8048. [PMID: 19956567 PMCID: PMC2779655 DOI: 10.1371/journal.pone.0008048] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 11/02/2009] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Sphingosine kinase-1 (SphK1) is an oncogenic lipid kinase notably involved in response to anticancer therapies in prostate cancer. Androgens regulate prostate cancer cell proliferation, and androgen deprivation therapy is the standard of care in the management of patients with advanced disease. Here, we explored the role of SphK1 in the regulation of androgen-dependent prostate cancer cell growth and survival. METHODOLOGY/PRINCIPAL FINDINGS Short-term androgen removal induced a rapid and transient SphK1 inhibition associated with a reduced cell growth in vitro and in vivo, an event that was not observed in the hormono-insensitive PC-3 cells. Supporting the critical role of SphK1 inhibition in the rapid effect of androgen depletion, its overexpression could impair the cell growth decrease. Similarly, the addition of dihydrotestosterone (DHT) to androgen-deprived LNCaP cells re-established cell proliferation, through an androgen receptor/PI3K/Akt dependent stimulation of SphK1, and inhibition of SphK1 could markedly impede the effects of DHT. Conversely, long-term removal of androgen support in LNCaP and C4-2B cells resulted in a progressive increase in SphK1 expression and activity throughout the progression to androgen-independence state, which was characterized by the acquisition of a neuroendocrine (NE)-like cell phenotype. Importantly, inhibition of the PI3K/Akt pathway--by negatively impacting SphK1 activity--could prevent NE differentiation in both cell models, an event that could be mimicked by SphK1 inhibitors. Fascinatingly, the reversability of the NE phenotype by exposure to normal medium was linked with a pronounced inhibition of SphK1 activity. CONCLUSIONS/SIGNIFICANCE We report the first evidence that androgen deprivation induces a differential effect on SphK1 activity in hormone-sensitive prostate cancer cell models. These results also suggest that SphK1 activation upon chronic androgen deprivation may serve as a compensatory mechanism allowing prostate cancer cells to survive in androgen-depleted environment, giving support to its inhibition as a potential therapeutic strategy to delay/prevent the transition to androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Audrey Dayon
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Leyre Brizuela
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Claire Martin
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Catherine Mazerolles
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- CHU Toulouse, Hôpital Rangueil, Laboratoire Anatomie Pathologique et Histologie-Cytologie, Toulouse, France
| | - Nelly Pirot
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Nicolas Doumerc
- Université de Toulouse, UPS, IPBS, Toulouse, France
- CHU Toulouse, Hôpital Rangueil, Service d'Urologie et de Transplantation Rénale, Toulouse, France
| | - Leonor Nogueira
- CHU Toulouse, Hôpital Purpan, Laboratoire de Biologie Cellulaire et Cytologie, Toulouse, France
| | - Muriel Golzio
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Justin Teissié
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Guy Serre
- CHU Toulouse, Hôpital Purpan, Laboratoire de Biologie Cellulaire et Cytologie, Toulouse, France
| | - Pascal Rischmann
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
- CHU Toulouse, Hôpital Rangueil, Service d'Urologie et de Transplantation Rénale, Toulouse, France
| | - Bernard Malavaud
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
- CHU Toulouse, Hôpital Rangueil, Service d'Urologie et de Transplantation Rénale, Toulouse, France
| | - Olivier Cuvillier
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, IPBS, Toulouse, France
- CHU Toulouse, Hôpital Rangueil, Service d'Urologie et de Transplantation Rénale, Toulouse, France
- * E-mail:
| |
Collapse
|
80
|
Grandoch M, Rose A, ter Braak M, Jendrossek V, Rübben H, Fischer JW, Schmidt M, Weber AA. Epac inhibits migration and proliferation of human prostate carcinoma cells. Br J Cancer 2009; 101:2038-42. [PMID: 19920825 PMCID: PMC2795436 DOI: 10.1038/sj.bjc.6605439] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: It was recently found that cAMP mediates protein kinase A-independent effects through Epac proteins. The aim of this study was to investigate the role of Epac in migration and proliferation of prostate carcinoma cells. Methods: The effect of Epac activation was determined by [3H]thymidine incorporation and scratch assays in PC-3 and DU 145 cells. Furthermore, cytoskeletal integrity was analysed by phalloidin staining. The participation of intracellular Epac effectors such as mitogen-activated protein (MAP) kinases, Rap1- and Rho-GTPases was determined by immunoblotting and pull-down assay. Results: The specific Epac activator 8-pCPT-2′-O-Me-cAMP (8-pCPT) interfered with cytoskeletal integrity, reduced DNA synthesis, and migration. Although 8-pCPT activated Rap1, it inhibited MAP kinase signalling and RhoA activation. These findings were translated into functional effects such as inhibition of mitogenesis, cytoskeletal integrity, and migration. Conclusion: In human prostate carcinoma cells, Epac inhibits proliferative and migratory responses likely because of inhibition of MAP kinase and RhoA signalling pathways. Therefore, Epac might represent an attractive therapeutic target in the treatment of prostate cancer.
Collapse
Affiliation(s)
- M Grandoch
- Department of Pharmacology, University of Essen Medical School, Essen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Rahrmann EP, Collier LS, Knutson TP, Doyal ME, Kuslak SL, Green LE, Malinowski RL, Roethe L, Akagi K, Waknitz M, Huang W, Largaespada DA, Marker PC. Identification of PDE4D as a proliferation promoting factor in prostate cancer using a Sleeping Beauty transposon-based somatic mutagenesis screen. Cancer Res 2009; 69:4388-97. [PMID: 19401450 PMCID: PMC2710962 DOI: 10.1158/0008-5472.can-08-3901] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Retroviral and transposon-based mutagenesis screens in mice have been useful for identifying candidate cancer genes for some tumor types. However, many of the organs that exhibit the highest cancer rates in humans, including the prostate, have not previously been amenable to these approaches. This study shows for the first time that the Sleeping Beauty transposon system can be used to identify candidate prostate cancer genes in mice. Somatic mobilization of a mutagenic transposon resulted in focal epithelial proliferation and hyperplasia in the prostate. Efficient methods were established to identify transposon insertion sites in these lesions, and analysis of transposon insertions identified candidate prostate cancer genes at common insertion sites, including Pde4d. PDE4D was also overexpressed in human prostate cancer patient samples and cell lines, and changes in PDE4D mRNA isoform expression were observed in human prostate cancers. Furthermore, knockdown of PDE4D reduced the growth and migration of prostate cancer cells in vitro, and knockdown of PDE4D reduced the growth and proliferation rate of prostate cancer xenografts in vivo. These data indicate that PDE4D functions as a proliferation promoting factor in prostate cancer, and the Sleeping Beauty transposon system is a useful tool for identifying candidate prostate cancer genes.
Collapse
Affiliation(s)
- Eric P. Rahrmann
- Department of Genetics, Cell Biology, and Development and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Lara S. Collier
- Division of Pharmaceutical Sciences, School of Pharmacy and UW Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Todd P. Knutson
- Department of Genetics, Cell Biology, and Development and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Meghan E. Doyal
- Department of Genetics, Cell Biology, and Development and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Sheri L. Kuslak
- Department of Genetics, Cell Biology, and Development and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Laura E. Green
- Department of Genetics, Cell Biology, and Development and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Rita L. Malinowski
- Division of Pharmaceutical Sciences, School of Pharmacy and UW Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Laura Roethe
- Division of Pharmaceutical Sciences, School of Pharmacy and UW Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Keiko Akagi
- Mouse Cancer Genetics Program, National Cancer Institute in Frederick, Frederick, MD, 21702, USA
| | - Michelle Waknitz
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - David A. Largaespada
- Department of Genetics, Cell Biology, and Development and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Paul C. Marker
- Division of Pharmaceutical Sciences, School of Pharmacy and UW Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| |
Collapse
|
82
|
Komiya A, Suzuki H, Imamoto T, Kamiya N, Nihei N, Naya Y, Ichikawa T, Fuse H. Neuroendocrine differentiation in the progression of prostate cancer. Int J Urol 2009; 16:37-44. [PMID: 19120524 DOI: 10.1111/j.1442-2042.2008.02175.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neuroendocrine (NE) cells originally exist in the normal prostate acini and duct, regulating prostatic growth, differentiation and secretion. Clusters of malignant NE cells are found in most prostate cancer (PCa) cases. NE differentiation (NED) is the basic character of the prostate, either benign or malignant. NE cells hold certain peptide hormones or pro-hormones, which affect the target cells by endocrine, paracrine, autocrine and neuroendocrine transmission in an androgen-independent fashion due to the lack of androgen receptor. NED is accessed by immunohistochemical staining or measurement of serum levels of NE markers. The extent of NED is associated with progression and prognosis of PCa. Chromogranin A (CGA) is the most important NE marker. In metastatic PCa, pretreatment serum CGA levels can be a predictor for progression and survival after endocrine therapy. It is recommended to measure longitudinal change in serum CGA. The NE pathway can also be a therapeutic target.
Collapse
Affiliation(s)
- Akira Komiya
- Department of Urology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Király M, Porcsalmy B, Pataki A, Kádár K, Jelitai M, Molnár B, Hermann P, Gera I, Grimm WD, Ganss B, Zsembery A, Varga G. Simultaneous PKC and cAMP activation induces differentiation of human dental pulp stem cells into functionally active neurons. Neurochem Int 2009; 55:323-32. [PMID: 19576521 DOI: 10.1016/j.neuint.2009.03.017] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 03/30/2009] [Indexed: 12/12/2022]
Abstract
The plasticity of dental pulp stem cells (DPSCs) has been demonstrated by several studies showing that they appear to self-maintain through several passages, giving rise to a variety of cells. The aim of the present study was to differentiate DPSCs to mature neuronal cells showing functional evidence of voltage gated ion channel activities in vitro. First, DPSC cultures were seeded on poly-l-lysine coated surfaces and pretreated for 48h with a medium containing basic fibroblast growth factor and the demethylating agent 5-azacytidine. Then neural induction was performed by the simultaneous activation of protein kinase C and the cyclic adenosine monophosphate pathway. Finally, maturation of the induced cells was achieved by continuous treatment with neurotrophin-3, dibutyryl cyclic AMP, and other supplementary components. Non-induced DPSCs already expressed vimentin, nestin, N-tubulin, neurogenin-2 and neurofilament-M. The inductive treatment resulted in decreased vimentin, nestin, N-tubulin and increased neurogenin-2, neuron-specific enolase, neurofilament-M and glial fibrillary acidic protein expression. By the end of the maturation period, all investigated genes were expressed at higher levels than in undifferentiated controls except vimentin and nestin. Patch clamp analysis revealed the functional activity of both voltage-dependent sodium and potassium channels in the differentiated cells. Our results demonstrate that although most surviving cells show neuronal morphology and express neuronal markers, there is a functional heterogeneity among the differentiated cells obtained by the in vitro differentiation protocol described herein. Nevertheless, this study clearly indicates that the dental pulp contains a cell population that is capable of neural commitment by our three step neuroinductive protocol.
Collapse
Affiliation(s)
- Marianna Király
- Department of Oral Biology, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Mushinski JF, Nguyen P, Stevens LM, Khanna C, Lee S, Chung EJ, Lee MJ, Kim YS, Linehan WM, Horisberger MA, Trepel JB. Inhibition of tumor cell motility by the interferon-inducible GTPase MxA. J Biol Chem 2009; 284:15206-14. [PMID: 19297326 DOI: 10.1074/jbc.m806324200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
To identify pathways controlling prostate cancer metastasis we performed differential display analysis of the human prostate carcinoma cell line PC-3 and its highly metastatic derivative PC-3M. This revealed that a 78-kDa interferon-inducible GTPase, MxA, was expressed in PC-3 but not in PC-3M cells. The gene encoding MxA, MX1, is located in the region of chromosome 21 deleted as a consequence of fusion of TMPRSS2 and ERG, which has been associated with aggressive, invasive prostate cancer. Stable exogenous MxA expression inhibited in vitro motility and invasiveness of PC-3M cells. In vivo exogenous MxA expression decreased the number of hepatic metastases following intrasplenic injection. Exogenous MxA also reduced motility and invasiveness of highly metastatic LOX melanoma cells. A mutation in MxA that inactivated its GTPase reversed inhibition of motility and invasion in both tumor cell lines. Co-immunoprecipitation studies demonstrated that MxA associated with tubulin, but the GTPase-inactivating mutation blocked this association. Because MxA is a highly inducible gene, an MxA-targeted drug discovery screen was initiated by placing the MxA promoter upstream of a luciferase reporter. Examination of the NCI diversity set of small molecules revealed three hits that activated the promoter. In PC-3M cells, these drugs induced MxA protein and inhibited motility. These data demonstrate that MxA inhibits tumor cell motility and invasion, and that MxA expression can be induced by small molecules, potentially offering a new approach to the prevention and treatment of metastasis.
Collapse
Affiliation(s)
- J Frederic Mushinski
- Laboratory of Cancer Biology and Genetics, Medical Oncology Branch, Pediatric Oncology Branch, and Urologic Oncology Branch, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Lara PN, Longmate J, Evans CP, Quinn DI, Twardowski P, Chatta G, Posadas E, Stadler W, Gandara DR. A phase II trial of the Src-kinase inhibitor AZD0530 in patients with advanced castration-resistant prostate cancer: a California Cancer Consortium study. Anticancer Drugs 2009; 20:179-84. [PMID: 19396016 PMCID: PMC3225398 DOI: 10.1097/cad.0b013e328325a867] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Prostate cancer cells undergo neuroendocrine differentiation during androgen deprivation and secrete neuropeptides, hence activating androgen receptor-regulated genes. Src-family protein kinases are involved in neuropeptide-induced prostate cancer growth and migration. A phase II trial of AZD0530, an oral Src-family kinase inhibitor, in patients with advanced castration resistant prostate cancer was conducted. The primary endpoint was prostate cancer-specific antigen (PSA) response rate, defined as a 30% or greater decrease. A two-stage Simon design was used. Eligibility criteria included documentation of castration resistance (including antiandrogen withdrawal), adequate end-organ function, and performance status, and not more than one prior taxane-based chemotherapy regimen. AZD0530 was given at 175 mg orally once daily continuously. Rapid accrual led to 28 patients registering in the first stage. Median age was 67 years. Sixteen patients had performance status (PS) 0, eight patients had PS 1, and four patients had PS 2. Nine patients (32%) had prior docetaxel-based chemotherapy. Five patients had transient PSA reductions not meeting PSA response criteria. Median progression-free survival time was 8 weeks. Treatment was generally well tolerated. AZD0530, a potent oral Src kinase inhibitor, is feasible and tolerable in this pretreated patient population but possessed little clinical efficacy as monotherapy. Strong preclinical evidence warrants further investigation of AZD0530 in earlier-stage prostate cancer or as combination therapy.
Collapse
Affiliation(s)
- Primo N Lara
- University of California Davis Cancer Center, Sacramento, California 95817, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Kung HJ, Evans CP. Oncogenic activation of androgen receptor. Urol Oncol 2009; 27:48-52. [PMID: 19111798 PMCID: PMC2629789 DOI: 10.1016/j.urolonc.2008.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 06/17/2008] [Accepted: 06/20/2008] [Indexed: 12/29/2022]
Abstract
BACKGROUND There is considerable evidence implicating the aberrant activation or "reactivation" of androgen receptor in the course of androgen-ablation therapy as a potential cause for the development of castration-resistant prostate cancer. Several non-mutually exclusive mechanisms including the inappropriate activation of androgen receptor (AR) by non-steroids have been postulated. The present work is aimed to understand the role of neuropeptides released by neuroendocrine transdifferentiated prostate cancer cells in the aberrant activation of AR. OBJECTIVES The study was designed to study how neuropeptides such as gastrin-releasing peptide activate AR and to define the crucial signal pathways involved, in the hope to identify therapeutic targets. METHODS AND MATERIALS Androgen-dependent LNCaP cell line was used to study the effects of bombesin/gastrin-releasing peptide on the growth of the cell line and the transactivation of AR. The neuropeptide was either added to the media or introduced as a transgene in LNCaP cells to study its paracrine or autocrine effect on LNCaP growth under androgen-deprived conditions. The activation of AR was monitored by reporter assay, chromatin immunoprecipitation (ChIP) of AR, translocation into the nucleus and cDNA microarray of the AR response genes. RESULTS Bombesin/gastrin releasing peptides induce androgen-independent growth of LNCaP in vitro and in vivo. It does so by activating AR, which is accompanied by the activation of Src tyrosine kinase and its target c-myc oncogene. The bombesin or Src-activated AR induces an overlapping set of AR response genes as androgen, but they also a unique set of genes. Intriguingly, the Src-activated and androgen-bound ARs differ in their binding specificity toward AR response elements, indicating the receptors activated by these 2 mechanisms are not conformationally identical. Finally, Src inhibitor was shown to effectively block the activation of AR and the growth effects induced by bombesin. CONCLUSION The results showed that AR can be activated by neuropeptide, a ligand for G-protein coupled receptor, in the absence of androgen. The activation goes through Src-tyrosine kinase pathway, and tyrosine kinase inhibitor is a potentially useful adjunctive therapy during androgen ablation.
Collapse
Affiliation(s)
- Hsing-Jien Kung
- Department of Basic Sciences, University of California, Davis Cancer Center, Sacramento, CA 95817, USA.
| | | |
Collapse
|
87
|
Sago K, Tamahara S, Tomihari M, Matsuki N, Asahara Y, Takei A, Bonkobara M, Washizu T, Ono K. In vitro differentiation of canine celiac adipose tissue-derived stromal cells into neuronal cells. J Vet Med Sci 2008; 70:353-7. [PMID: 18460829 DOI: 10.1292/jvms.70.353] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To investigate in vitro differentiation of canine adipose tissue-derived stromal cells (ATSCs) into neuronal cells, ATSCs from celiac adipose tissue in clinically healthy beagle dogs were treated with 100 muM dibutyryl cyclic adenosine monophosphate (dbcAMP) and 125 muM isobuthylmethylxanthine (IBMX). ATSCs were morphologically changed into differentiated ATSCs from spindle-shaped cells to neuron-like cells with numerous processes after the treatment. Expression of neuron-specific enolase (NSE) as an early neuron specific marker protein was detected in both ATSCs and differentiated ATSCs, however diachronic increase of NSE expression was observed in differentiated ATSCs after the treatment with dbcAMP/IBMX. In addition, neurofilament-68 (NF-68) as an early to mature neuron specific marker protein was weakly expressed in differentiated ATSCs. Neuron specific glutamate and glucose transporter (EAAC1 and GLUT-3, respectively) mRNAs were strongly expressed in differentiated ATSCs compared with those in ATSCs, although glia specific glutamate transporter mRNA (GLT-1) was also detected in differentiated ATSCs. ATSCs can differentiate into early to mature neuronal cells and are candidate cells for autologous nerve regeneration therapy, although additional research is needed to examine functional characteristics of differentiated ATSCs.
Collapse
Affiliation(s)
- Ken Sago
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Chang YM, Bai L, Liu S, Yang JC, Kung HJ, Evans CP. Src family kinase oncogenic potential and pathways in prostate cancer as revealed by AZD0530. Oncogene 2008; 27:6365-75. [PMID: 18679417 PMCID: PMC4294546 DOI: 10.1038/onc.2008.250] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 06/03/2008] [Accepted: 06/10/2008] [Indexed: 12/16/2022]
Abstract
Prostate cancer is the most frequently diagnosed cancer in American men. We have previously demonstrated that Src mediates androgen-independent proliferation in prostate cancer. We sought to investigate the Src-mediated oncogenic pathways and tumor biology using AZD0530, a novel Src family kinase/Abl dual-kinase inhibitor that is entering phase II clinical trials. We show that while both Src and Abl are expressed in all prostate cancer cell lines, Src but not Abl is activated in the prostate. Furthermore, Src activation is inhibited by AZD0530 in a rapid and dose-dependent manner. We show that Src mediates cell proliferation in DU145 and PC3 cells at the G1 phase of cell cycle. Src inhibition resulted in decreased binding of beta-catenin to the promoters of G1 phase cell cycle regulators cyclin D1 and c-Myc. C-Myc may also be regulated at the protein level by extracellular signal-regulated kinase 1/2 and GSK3beta. Cell motility factors focal adhesion kinase, p130CAS and paxillin activation in DU145 and PC3 cells were also inhibited. Administration of AZD0530 in mice reduced orthotopic DU145 xenograft growth by 45%. We have further delineated the Src-mediated oncogenic growth and migration pathways in prostate cancer and established mechanistic rationale for Src inhibition as novel therapy in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Y-M Chang
- Department of Urology, University of California at Davis, Sacramento, CA, USA
| | - L Bai
- Department of Urology, University of California at Davis, Sacramento, CA, USA
| | - S Liu
- Department of Biological Chemistry and Molecular Medicine, University of California at Davis, Sacramento, CA, USA
| | - JC Yang
- Department of Urology, University of California at Davis, Sacramento, CA, USA
| | - H-J Kung
- Department of Urology, University of California at Davis, Sacramento, CA, USA
- Department of Biological Chemistry and Molecular Medicine, University of California at Davis, Sacramento, CA, USA
- Cancer Center, University of California at Davis, Sacramento, CA, USA
| | - CP Evans
- Department of Urology, University of California at Davis, Sacramento, CA, USA
- Cancer Center, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
89
|
Gulubova M, Vlaykova T. Chromogranin A-, serotonin-, synaptophysin- and vascular endothelial growth factor-positive endocrine cells and the prognosis of colorectal cancer: an immunohistochemical and ultrastructural study. J Gastroenterol Hepatol 2008; 23:1574-85. [PMID: 18771509 DOI: 10.1111/j.1440-1746.2008.05560.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Endocrine differentiation in colorectal adenocarcinoma has been reported but its significance as a prognostic marker remains uncertain. The aim of the present study was to analyze the prognostic significance of endocrine differentiation in colorectal cancer. METHODS The presence of endocrine cells (EC) was determined in 137 colorectal cancers using light and electron immunohistochemistry and the immunogold method with chromogranin A, serotonin and synaptophysin. Vascular endothelial growth factor (VEGF) expression in tumor biopsies was also analyzed applying anti-VEGF antibodies. RESULTS EC labeled with at least one of the studied markers were detected in 47 (34.3%) primary colorectal cancers (30% chromogranin A-positive, 33% synaptophysin-positive and 18% serotonin-positive). In 23% of tumor biopsies, VEGF-positive EC were also detected. The immunostaining on serial sections showed that some chromogranin A-, synaptophysin- or serotonin-positive EC also contained VEGF immune deposits. By the immunogold method, the presence of VEGF was localized to the granules of EC. Tumors with VEGF-positive EC appeared to have significantly higher vascularization, detected as systematic microvessel density (28.89 vs 15.22 vessels/mm(2), P = 0.044, Mann-Whitney U-test) compared to those without VEGF-positive EC. Ultrastructurally, EC in the tumor tissue displayed some features different from those in the normal colon. The survival analyses revealed that patients with EC in primary tumor tissues had a worse prognosis after surgical therapy than those without endocrine cell differentiation (P < 0.05, log-rank test). CONCLUSIONS Endocrine differentiation is not an uncommon event in primary colorectal cancer and it could be a useful marker for a worse prognosis after the surgical therapy. Tumors positive for VEGF and containing VEGF-positive EC have higher vascularization, which probably also contributes to the unfavorable prognosis of patients.
Collapse
Affiliation(s)
- Maya Gulubova
- Department of General and Clinical Pathology, Medical Faculty, Trakia University, Stara Zagora, Bulgaria.
| | | |
Collapse
|
90
|
Frigo DE, McDonnell DP. Differential effects of prostate cancer therapeutics on neuroendocrine transdifferentiation. Mol Cancer Ther 2008; 7:659-69. [PMID: 18347151 DOI: 10.1158/1535-7163.mct-07-0480] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Androgen ablation therapy is widely used for the treatment of advanced prostate cancer. However, the effectiveness of this intervention strategy is generally short-lived as the disease ultimately progresses to a hormone-refractory state. In recent years, it has become clear that even in antiandrogen-resistant cancers the androgen receptor (AR) signaling axis is intact and is required for prostate cancer growth. Thus, there is a heightened interest in developing small molecules that function in part by down-regulating AR expression in tumors. Paradoxically, AR expression has been shown to be important in preventing the transdifferentiation of epithelial prostate cancer cells toward a neuroendocrine phenotype associated with tumor progression. Consequently, we have evaluated the relative effect of prostate cancer therapeutics that function in part by depleting AR levels on neuroendocrine differentiation in established cellular models of prostate cancer. These studies reveal that although histone deacetylase inhibitors can down-regulate AR expression they increase the expression of neuroendocrine markers and alter cellular morphology. Inhibition of AR signaling using classic AR antagonists or small interfering RNA-mediated AR ablation induces incomplete neuroendocrine differentiation. Importantly, the Hsp90 inhibitor geldanamycin effectively down-regulates AR expression while having no effect on neuroendocrine differentiation. Taken together, these data show that the phenotypic responses to pharmacologic agents used in the clinic to prevent the progression of prostate cancer are not equivalent, a finding of significant therapeutic importance.
Collapse
Affiliation(s)
- Daniel E Frigo
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
91
|
Gackière F, Bidaux G, Delcourt P, Van Coppenolle F, Katsogiannou M, Dewailly E, Bavencoffe A, Van Chuoï-Mariot MT, Mauroy B, Prevarskaya N, Mariot P. CaV3.2 T-type calcium channels are involved in calcium-dependent secretion of neuroendocrine prostate cancer cells. J Biol Chem 2008; 283:10162-73. [PMID: 18230611 DOI: 10.1074/jbc.m707159200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Because prostate cancer is, in its early stages, an androgen-dependent pathology, treatments aiming at decreasing testosterone plasma concentration have been developed for many years now. However, a significant proportion of patients suffer a relapse after a few years of hormone therapy. The androgen-independent stage of prostate cancer has been shown to be associated with the development of neuroendocrine differentiation. We previously demonstrated that neuroendocrine prostate cancer cells derived from LNCaP cells overexpress CaV3.2 T-type voltage-dependent calcium channels. We demonstrate here using prostatic acid phosphatase as a marker of prostate secretion and FM1-43 fluorescence imaging of membrane trafficking that neuroendocrine differentiation is associated with an increase in calcium-dependent secretion which critically relies on CaV3.2 T-type calcium channel activity. In addition, we show that these channels are expressed by neuroendocrine cells in prostate cancer tissues obtained from patients after surgery. We propose that CaV3.2 T-type calcium channel up-regulation may account for the alteration of secretion during prostate cancer development and that these channels, by promoting the secretion of potential mitogenic factors, could participate in the progression of the disease toward an androgen-independent stage.
Collapse
Affiliation(s)
- Florian Gackière
- INSERM U800, Laboratoire de Physiologie Cellulaire, Equipe Labellisée par la Ligue contre le Cancer and Université des Sciences et Technologies de Lille, Villeneuve d'Ascq, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Origin of androgen-insensitive poorly differentiated tumors in the transgenic adenocarcinoma of mouse prostate model. Neoplasia 2007; 9:938-50. [PMID: 18030362 DOI: 10.1593/neo.07562] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Revised: 09/13/2007] [Accepted: 09/17/2007] [Indexed: 11/18/2022] Open
Abstract
Following castration, the transgenic adenocarcinoma of mouse prostate (TRAMP) model demonstrates rapid development of SV40-Tag-driven poorly differentiated tumors that express neuroendocrine cell markers. The cell population dynamics within the prostates of castrated TRAMP mice were characterized by analyzing the incorporation of 5-bromodeoxyuridine (BrdUrd) and the expression of SV40-Tag, synaptophysin, and androgen receptor (AR). Fourteen days postcastration, the remaining epithelial cells and adenocarcinoma cells were nonproliferative and lacked detectable SV40-Tag or synaptophysin expression. In contrast, morphologically distinct intraglandular foci were identified which expressed SV40-Tag, synaptophysin, and Ki67, but that lacked AR expression. These proliferative SV40-Tag and synaptophysin-expressing intraglandular foci were associated with the rare BrdUrd-retaining cells. These foci expanded rapidly in the postcastration prostate environment, in contrast to the AR- and SV40-Tag-expressing adenocarcinoma cells that lost SV40-Tag expression and underwent apoptosis after castration. Intraglandular foci of synaptophysin-expressing cells were also observed in the prostates of intact TRAMP mice at a comparable frequency; however, they did not progress to rapidly expanding tumors until much later in the life of the mice. This suggests that the foci of neuroendocrine-like cells that express SV40-Tag and synaptophysin, but lack AR, arise independent of androgen-deprivation and represent the source of the poorly differentiated tumors that are the lethal phenotype in the TRAMP model.
Collapse
|
93
|
Martín-Orozco RM, Almaraz-Pro C, Rodríguez-Ubreva FJ, Cortés MA, Ropero S, Colomer R, López-Ruiz P, Colás B. EGF prevents the neuroendocrine differentiation of LNCaP cells induced by serum deprivation: the modulator role of PI3K/Akt. Neoplasia 2007; 9:614-24. [PMID: 17898861 PMCID: PMC1950431 DOI: 10.1593/neo.07337] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 06/06/2007] [Accepted: 06/07/2007] [Indexed: 11/18/2022]
Abstract
The primary focus of this investigation was to study the relationship between neuroendocrine (NE) differentiation and epidermal growth factor (EGF) because both have been implicated in the progression of prostate cancer. For this purpose, we used gefitinib and trastuzumab, which are inhibitors of EGF receptor (EGFR) and ErbB2, respectively. EGF prevents NE differentiation induced by androgen depletion. This effect is prevented by gefitinib, which blocks the activation of EGFR and ErbB2, stimulation of mitogen-activated protein kinase (MAPK), and cell proliferation induced by EGF. Conversely, trastuzumab does not inhibit the effect of EGF on EGFR phosphorylation, MAPK activity, cell proliferation, and NE differentiation, although it reduces ErbB2 levels specifically, suggesting that ErbB2 is not necessary to inhibit NE differentiation. Prevention of NE differentiation by EGF is mediated by a MAPK-dependent mechanism and requires constitutive Akt activation. The abrogation of the PI3K/Akt pathway changes the role of EGF from inhibitor to inductor of NE differentiation. We show that EGFR tyrosine kinase, MAPK, and PI3K inhibitors inhibit the cell proliferation stimulated by EGF but induce the acquisition of NE phenotype. Altogether, the present data should be borne in mind when designing new clinical schedules for the treatment of prostate cancer, including the use of ErbB receptors and associated signaling pathway inhibitors.
Collapse
Affiliation(s)
- Rosa M Martín-Orozco
- Department of Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, Madrid E-28871, Spain
| | - Carmén Almaraz-Pro
- Department of Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, Madrid E-28871, Spain
| | - F Javier Rodríguez-Ubreva
- Department of Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, Madrid E-28871, Spain
| | - M Alicia Cortés
- Department of Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, Madrid E-28871, Spain
| | - Santiago Ropero
- Epigenetics Laboratory, Spanish National Cancer Center, Melchor Fernandez Almagro 3, Madrid 28029, Spain
| | - Ramón Colomer
- Medical Oncology, Institut Catala Oncologia, Hospital Universitari de Girona Dr. Josep Trueta, Girona, Spain
| | - Pilar López-Ruiz
- Department of Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, Madrid E-28871, Spain
| | - Begoña Colás
- Department of Bioquímica y Biología Molecular, Universidad de Alcalá, Alcalá de Henares, Madrid E-28871, Spain
| |
Collapse
|
94
|
Deeble PD, Cox ME, Frierson HF, Sikes RA, Palmer JB, Davidson RJ, Casarez EV, Amorino GP, Parsons SJ. Androgen-independent growth and tumorigenesis of prostate cancer cells are enhanced by the presence of PKA-differentiated neuroendocrine cells. Cancer Res 2007; 67:3663-72. [PMID: 17440078 DOI: 10.1158/0008-5472.can-06-2616] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The neuroendocrine status of prostatic adenocarcinomas is considered a prognostic indicator for development of aggressive, androgen-independent disease. Neuroendocrine-like cells are thought to function by providing growth and survival signals to surrounding tumor cells, particularly following androgen ablation therapy. To test this hypothesis directly, LNCaP cells were engineered to inducibly express a constitutively activated form of the cyclic AMP-dependent protein kinase A catalytic subunit (caPKA), which was previously found upon transient transfection to be sufficient for acquisition of neuroendocrine-like characteristics and loss of mitotic activity. Clonal cells that inducibly expressed caPKA enhanced the growth of prostate tumor cells in anchorage-dependent and anchorage-independent in vitro assays as well as the growth of prostate tumor xenografts in vivo, with the greatest effects seen under conditions of androgen deprivation. These results suggest that neuroendocrine-like cells of prostatic tumors have the potential to enhance androgen-independent tumor growth in a paracrine manner, thereby contributing to progression of the disease.
Collapse
Affiliation(s)
- Paul D Deeble
- Department of Microbiology and Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Shimizu S, Kumagai J, Eishi Y, Uehara T, Kawakami S, Takizawa T, Koike M. Frequency and number of neuroendocrine tumor cells in prostate cancer: no difference between radical prostatectomy specimens from patients with and without neoadjuvant hormonal therapy. Prostate 2007; 67:645-52. [PMID: 17342745 DOI: 10.1002/pros.20493] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Neuroendocrine tumor cells in prostate cancer are thought to increase after hormonal therapy due to neuroendocrine differentiation of tumor cells. This assumption is based on the histological analyses of limited portions of the cancerous lesions examined. METHODS Radical prostatectomy specimens were obtained from 122 consecutive patients with prostate adenocarcinoma, 70 of whom underwent prostatectomy alone (Group A) and 52 with neoadjuvant hormonal therapy (Group B). Sections from all the 5-mm-thick slices from formalin-fixed specimens were immunostained for chromogranin-A, and the total number of choromogranin-A-positive neuroendocrine tumor cells were counted. RESULTS No difference was found between Groups A and B in the frequency of cancer with neuroendocrine cells. The total number of neuroendocrine cells in cancer varied widely with no difference of median values in the two groups. CONCLUSIONS These results do not support the assumption that hormonal therapy induces neuroendocrine differentiation, but suggest androgen-independent neuroendocrine cells existed before therapy.
Collapse
Affiliation(s)
- Shinichiro Shimizu
- Department of Human Pathology, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
BACKGROUND Carcinoma of the prostate (CaP) is the most commonly diagnosed cancer in men in the United States. Signal transduction molecules such as tyrosine kinases play important roles in CaP. Src, a nonreceptor tyrosine kinase (NRTK) and the first proto-oncogene discovered is shown to participate in processes such as cell proliferation and migration in CaP. Underscoring NRTK's and, specifically, Src's importance in cancer is the recent approval by the US Food and Drug Administration of dasatinib, the first commercial Src inhibitor for clinical use in chronic myelogenous leukemia (CML). In this review we will focus on NRTKs and their roles in the biology of CaP. MATERIALS AND METHODS Publicly available literature from PubMed regarding the topic of members of NRTKs in CaP was searched and reviewed. RESULTS Src, FAK, JaK1/2, and ETK are involved in processes indispensable to the biology of CaP: cell growth, migration, invasion, angiogenesis, and apoptosis. CONCLUSIONS Src emerges as a common signaling and regulatory molecule in multiple biological processes in CaP. Src's relative importance in particular stages of CaP, however, required further definition. Continued investigation of NRTKs will increase our understanding of their biological function and potential role as new therapeutic targets.
Collapse
Affiliation(s)
- Yu-Ming Chang
- Department of Urology, University of California at Davis, Sacramento, CA 95817, USA
| | | | | |
Collapse
|
97
|
Kvissel AK, Ramberg H, Eide T, Svindland A, Skålhegg BS, Taskén KA. Androgen dependent regulation of protein kinase A subunits in prostate cancer cells. Cell Signal 2007; 19:401-9. [PMID: 16949795 DOI: 10.1016/j.cellsig.2006.07.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Revised: 07/11/2006] [Accepted: 07/18/2006] [Indexed: 11/28/2022]
Abstract
Neuroendocrine (NE) cells may play a role in prostate cancer progression. Both androgen deprivation and cAMP are well known inducers of NE differentiation (NED) in the prostate. Gene-expression profiling of LNCaP cells, incubated in androgen stripped medium, showed that the Cbeta isoform of PKA is up-regulated during NE differentiation. Furthermore, by using semi-quantitative RT-PCR and immunoblotting analysis, we observed that the Cbeta splice variants are differentially regulated during this process. Whereas the Cbeta2 splice variant is down-regulated in growth arrested LNCaP cells, the Cbeta1, Cbeta3 and Cbeta4 variants, as well as the RIIbeta subunit of PKA, are induced in NE-like LNCaP cells. The opposite effect of Cbeta expression could be mimicked by androgen stimulation, implying the Cbeta gene of PKA as a putative new target gene for the androgen receptor in prostate cancer. Moreover, to investigate expression of PKA subunits during prostate cancer progression, we did immunoblotting of several prostatic cell lines and normal and tumor tissue from prostate cancer patients. Interestingly, multiple Cbeta subunits were also observed in human prostate specimens, and the Cbeta2 variant was up-regulated in tumor cells. In conclusion, it seems that the Cbeta isoforms play different roles in proliferation and differentiation and could therefore be potential markers for prostate cancer progression.
Collapse
|
98
|
Ückert S, Hedlund P, Andersson KE, Truss MC, Jonas U, Stief CG. Update on Phosphodiesterase (PDE) Isoenzymes as Pharmacologic Targets in Urology: Present and Future. Eur Urol 2006; 50:1194-207; discussion 1207. [DOI: 10.1016/j.eururo.2006.05.025] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2004] [Accepted: 05/08/2006] [Indexed: 01/23/2023]
|
99
|
Nelson EC, Cambio AJ, Yang JC, Ok JH, Lara PN, Evans CP. Clinical implications of neuroendocrine differentiation in prostate cancer. Prostate Cancer Prostatic Dis 2006; 10:6-14. [PMID: 17075603 DOI: 10.1038/sj.pcan.4500922] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The cellular signaling pathways of the prostate play a central role in the induction, maintenance, and progression of prostate cancer (CaP). Neuroendocrine (NE) cells demonstrate attributes that suggest they are an integral part of these signaling cascades. We summarize what is known regarding NE cells in CaP focusing on NE cellular transdifferentiation. This significant event in CaP progression appears to be accelerated by androgen deprivation (AD) treatment. We examine biochemical pathways that may impact NE differentiation in a chronological manner focusing on AD therapy (ADT) as a central event in inducing androgen-independent CaP. Our analysis is limited to the common adenocarcinoma pattern of CaP and excludes small-cell and carcinoid prostatic variants. In conclusion, we speculate on the future of treatment and research in this area.
Collapse
Affiliation(s)
- E C Nelson
- Department of Urology, Davis Medical Center, University of California at Davis, Sacramento, CA 95817, USA
| | | | | | | | | | | |
Collapse
|
100
|
Ketata S, Ketata H, Fakhfakh H, Sahnoun A, Bahloul A, Boudawara T, Mhiri MN. Pure primary neuroendocrine tumor of the prostate: a rare entity. Clin Genitourin Cancer 2006; 5:82-4. [PMID: 16859584 DOI: 10.3816/cgc.2006.n.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
An extrapulmonary neuroendocrine tumor arising in the prostate gland has been described in several case series; however, pure and primary prostatic neuroendocrine tumors are very rare, their biologic behavior is not yet well known, and there are only a few reports in the literature describing the clinical features and management of this neoplasm. This article presents the clinicopathologic findings of 2 patients presenting with this disease. The published reports of primary pure neuroendocrine prostate tumors are discussed.
Collapse
Affiliation(s)
- Sabeur Ketata
- Department of Urology, CHU Habib Bourguiba, Rue Majida Boulila, Sfax, Tunisia.
| | | | | | | | | | | | | |
Collapse
|