101
|
Abstract
Multiple studies have confirmed that speckle-type pox virus and zinc finger (POZ) protein (SPOP) functions as a substrate adaptor of cullin 3-based E3 ligase and has a crucial role in various cellular processes via specific targeting of proteins for ubiquitination and subsequent proteasomal degradation. Dysregulation of SPOP-mediated proteolysis might be involved in the development and progression of human prostate and kidney cancers. In prostate cancer, SPOP seems to function as a tumour suppressor by targeting several proteins, including androgen receptor (AR), steroid receptor coactivator 3 (SRC3) and BRD4, for degradation, whereas it might function as an oncoprotein in kidney cancer, for example, by targeting phosphatase and tensin homologue (PTEN) for proteasomal degradation. In addition, nuclear SPOP targets AR for degradation and has a role as a tumour suppressor in prostate cancer; however, in kidney cancer, SPOP largely accumulates in the cytoplasm and fails to promote degradation of AR located in the nucleus, resulting in activation of AR-driven pathways and cancer progression. Owing to the context-dependent function of SPOP in human malignancies, further assessment of the molecular mechanisms involving SPOP in prostate and kidney cancers is needed to improve our understanding of its role in the development of these cancer types. Treatments that target SPOP might become therapeutic strategies in these malignancies in the future.
Collapse
|
102
|
Cui X, Piao C, Lv C, Lin X, Zhang Z, Liu X. ZNFX1 anti-sense RNA 1 promotes the tumorigenesis of prostate cancer by regulating c-Myc expression via a regulatory network of competing endogenous RNAs. Cell Mol Life Sci 2020; 77:1135-1152. [PMID: 31321444 PMCID: PMC11104963 DOI: 10.1007/s00018-019-03226-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 07/05/2019] [Accepted: 07/09/2019] [Indexed: 12/24/2022]
Abstract
ZNFX1 anti-sense RNA 1 (ZFAS1) has been indicated in the tumorigenesis of various human cancers. However, the role of ZFAS1 in prostate cancer (PCa) progression and the underlying mechanisms remain incompletely understood. In the present study, we discovered that ZFAS1 is upregulated in PCa and that ZFAS1 overexpression predicted poor clinical outcomes. ZFAS1 overexpression notably promoted the proliferation, invasion, and epithelial-mesenchymal transition of PCa cells. Furthermore, we not only discovered that miR-27a/15a/16 are targeted by ZFAS1, which binds to their miRNA-response elements, but also revealed their tumor suppressor roles in PCa. We also identified that the Hippo pathway transducer YAP1, as well as its cooperator, TEAD1, are common downstream targets of miR-27a/15a/16. In addition, H3K9 demethylase KDM3A was found to be another target gene of miR-27a. Importantly, YAP1, TEAD1, and KDM3A all act as strong c-Myc inducers in an androgen-independent manner. Taken together, we suggest a regulatory network in which ZFAS1 is capable of enhancing c-Myc expression by inducing the expression of YAP1, TEAD1, and KDM3A through crosstalk with their upstream miRNAs, thereby globally promoting prostate cancer tumorigenesis.
Collapse
Affiliation(s)
- Xiaolu Cui
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Chiyuan Piao
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Chengcheng Lv
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, China
| | - Xuyong Lin
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, 110001, China
| | - Zhe Zhang
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiankui Liu
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
103
|
Ki67 in Gleason Pattern 3 as a Marker of the Presence of Higher-Grade Prostate Cancer. Appl Immunohistochem Mol Morphol 2020; 29:112-117. [DOI: 10.1097/pai.0000000000000835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 01/12/2020] [Indexed: 11/26/2022]
|
104
|
Caggiano C, Pieraccioli M, Panzeri V, Sette C, Bielli P. c-MYC empowers transcription and productive splicing of the oncogenic splicing factor Sam68 in cancer. Nucleic Acids Res 2020; 47:6160-6171. [PMID: 31066450 PMCID: PMC6614821 DOI: 10.1093/nar/gkz344] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
The splicing factor Sam68 is upregulated in many human cancers, including prostate cancer (PCa) where it promotes cell proliferation and survival. Nevertheless, in spite of its frequent upregulation in cancer, the mechanism(s) underlying its expression are largely unknown. Herein, bioinformatics analyses identified the promoter region of the Sam68 gene (KHDRBS1) and the proto-oncogenic transcription factor c-MYC as a key regulator of Sam68 expression. Upregulation of Sam68 and c-MYC correlate in PCa patients. c-MYC directly binds to and activates the Sam68 promoter. Furthermore, c-MYC affects productive splicing of the nascent Sam68 transcript by modulating the transcriptional elongation rate within the gene. Importantly, c-MYC-dependent expression of Sam68 is under the tight control of external cues, such as androgens and/or mitogens. These findings uncover an unexpected coordination of transcription and splicing of Sam68 by c-MYC, which may represent a key step in PCa tumorigenesis.
Collapse
Affiliation(s)
- Cinzia Caggiano
- Laboratory of Neuroembryology, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Marco Pieraccioli
- Laboratory of Neuroembryology, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Valentina Panzeri
- Laboratory of Neuroembryology, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy.,Department of Science medical/chirurgic and translational medicine, University of Rome Sapienza,00189 Rome, Italy
| | - Claudio Sette
- Laboratory of Neuroembryology, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy.,Institute of Human Anatomy and Cell Biology, Catholic University of the Sacred Hearth, 00168 Rome, Italy
| | - Pamela Bielli
- Laboratory of Neuroembryology, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy.,Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
105
|
Pathway-guided analysis identifies Myc-dependent alternative pre-mRNA splicing in aggressive prostate cancers. Proc Natl Acad Sci U S A 2020; 117:5269-5279. [PMID: 32086391 PMCID: PMC7071906 DOI: 10.1073/pnas.1915975117] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We sought to define the landscape of alternative pre-mRNA splicing in prostate cancers and the relationship of exon choice to known cancer driver alterations. To do so, we compiled a metadataset composed of 876 RNA-sequencing (RNA-Seq) samples from five publicly available sources representing a range of prostate phenotypes from normal tissue to drug-resistant metastases. We subjected these samples to exon-level analysis with rMATS-turbo, purpose-built software designed for large-scale analyses of splicing, and identified 13,149 high-confidence cassette exon events with variable incorporation across samples. We then developed a computational framework, pathway enrichment-guided activity study of alternative splicing (PEGASAS), to correlate transcriptional signatures of 50 different cancer driver pathways with these alternative splicing events. We discovered that Myc signaling was correlated with incorporation of a set of 1,039 cassette exons enriched in genes encoding RNA binding proteins. Using a human prostate epithelial transformation assay, we confirmed the Myc regulation of 147 of these exons, many of which introduced frameshifts or encoded premature stop codons. Our results connect changes in alternative pre-mRNA splicing to oncogenic alterations common in prostate and many other cancers. We also establish a role for Myc in regulating RNA splicing by controlling the incorporation of nonsense-mediated decay-determinant exons in genes encoding RNA binding proteins.
Collapse
|
106
|
TIP5 primes prostate luminal cells for the oncogenic transformation mediated by PTEN-loss. Proc Natl Acad Sci U S A 2020; 117:3637-3647. [PMID: 32024754 PMCID: PMC7035629 DOI: 10.1073/pnas.1911673117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The cell of origin and the temporal order of oncogenic events in tumors play important roles for disease state. This is of particular interest for PCa due to its highly variable clinical outcome. However, these features are difficult to analyze in tumors. We established an in vitro murine PCa organoid model taking into account the cell of origin and the temporal order of events. We found that TIP5 primes luminal prostate cells for Pten-loss mediated oncogenic transformation whereas it is dispensable once the transformation is established. Cross-species transcriptomic analyses revealed a PTEN-loss gene signature that identified a set of aggressive tumors with PTEN-del, or low PTEN expression, and high-TIP5 expression. This paper provides a powerful tool to elucidate PCa mechanisms. Prostate cancer (PCa) is the second leading cause of cancer death in men. Its clinical and molecular heterogeneities and the lack of in vitro models outline the complexity of PCa in the clinical and research settings. We established an in vitro mouse PCa model based on organoid technology that takes into account the cell of origin and the order of events. Primary PCa with deletion of the tumor suppressor gene PTEN (PTEN-del) can be modeled through Pten-down-regulation in mouse organoids. We used this system to elucidate the contribution of TIP5 in PCa initiation, a chromatin regulator that is implicated in aggressive PCa. High TIP5 expression correlates with primary PTEN-del PCa and this combination strongly associates with reduced prostate-specific antigen (PSA) recurrence-free survival. TIP5 is critical for the initiation of PCa of luminal origin mediated by Pten-loss whereas it is dispensable once Pten-loss mediated transformation is established. Cross-species analyses revealed a PTEN gene signature that identified a group of aggressive primary PCas characterized by PTEN-del, high-TIP5 expression, and a TIP5-regulated gene expression profile. The results highlight the modeling of PCa with organoids as a powerful tool to elucidate the role of genetic alterations found in recent studies in their time orders and cells of origin, thereby providing further optimization for tumor stratification to improve the clinical management of PCa.
Collapse
|
107
|
Umemori M, Kurata M, Yamamoto A, Yamamoto K, Ishibashi S, Ikeda M, Tashiro K, Kimura T, Sato S, Takahashi H, Kitagawa M. The expression of MYC is strongly dependent on the circular PVT1 expression in pure Gleason pattern 4 of prostatic cancer. Med Mol Morphol 2020; 53:156-167. [PMID: 31932969 DOI: 10.1007/s00795-020-00243-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/03/2020] [Indexed: 12/31/2022]
Abstract
PVT1 is a long-noncoding RNA and is highly expressed in various cancers including prostate cancers with stabilizing MYC protein. To characterize the objective biological features of the different morphological components such as Gleason patterns (GP) in prostate cancer, biopsy specimens containing only single pure GP (GP3, GP4, GP5) are used to analyze the relationship between PVT1 expression and MYC protein expression. The expressions of PVT1 and MYC were analyzed by quantitative PCR and the labeling index (LI) of MYC protein by immunohistochemical staining. PVT1, MYC, and MYC protein were highly expressed in GP 4, and interestingly the expression between PVT1 and MYC LI significantly correlated only in GP 4. In vitro experiments, the expression of MYC protein was slightly reduced by small interfering RNA against PVT1, while strongly reduced against specifically circular PVT1, splicing variants derived from the PVT1. Taken together, the expression characteristics of PVT1, MYC, and MYC protein differed depending on the GP. In particular, circular PVT1 might be strongly involved in the stabilization of MYC protein in GP4 and suggest different biological features.
Collapse
Affiliation(s)
- Miyaka Umemori
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.,Department of Pathology, The Jikei University Hospital, Tokyo, Japan
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Akiko Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Sachiko Ishibashi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Masumi Ikeda
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kojiro Tashiro
- Department of Urology, The Jikei University Hospital, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University Hospital, Tokyo, Japan
| | - Shun Sato
- Department of Pathology, The Jikei University Hospital, Tokyo, Japan
| | | | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
108
|
Li Q, Liu B, Chao HP, Ji Y, Lu Y, Mehmood R, Jeter C, Chen T, Moore JR, Li W, Liu C, Rycaj K, Tracz A, Kirk J, Calhoun-Davis T, Xiong J, Deng Q, Huang J, Foster BA, Gokhale A, Chen X, Tang DG. LRIG1 is a pleiotropic androgen receptor-regulated feedback tumor suppressor in prostate cancer. Nat Commun 2019; 10:5494. [PMID: 31792211 PMCID: PMC6889295 DOI: 10.1038/s41467-019-13532-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
LRIG1 has been reported to be a tumor suppressor in gastrointestinal tract and epidermis. However, little is known about the expression, regulation and biological functions of LRIG1 in prostate cancer (PCa). We find that LRIG1 is overexpressed in PCa, but its expression correlates with better patient survival. Functional studies reveal strong tumor-suppressive functions of LRIG1 in both AR+ and AR- xenograft models, and transgenic expression of LRIG1 inhibits tumor development in Hi-Myc and TRAMP models. LRIG1 also inhibits castration-resistant PCa and exhibits therapeutic efficacy in pre-established tumors. We further show that 1) AR directly transactivates LRIG1 through binding to several AR-binding sites in LRIG1 locus, and 2) LRIG1 dampens ERBB expression in a cell type-dependent manner and inhibits ERBB2-driven tumor growth. Collectively, our study indicates that LRIG1 represents a pleiotropic AR-regulated feedback tumor suppressor that functions to restrict oncogenic signaling from AR, Myc, ERBBs, and, likely, other oncogenic drivers.
Collapse
Affiliation(s)
- Qiuhui Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, 430079, Wuhan, China
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Bigang Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Hsueh-Ping Chao
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Yibing Ji
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Rashid Mehmood
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Collene Jeter
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Taiping Chen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - John R Moore
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Wenqian Li
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Can Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Kiera Rycaj
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Amanda Tracz
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jason Kirk
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tammy Calhoun-Davis
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Jie Xiong
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Qu Deng
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University of School of Medicine, Durham, NC, 27710, USA
| | - Barbara A Foster
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Abhiram Gokhale
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Xin Chen
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA.
- Department of Oncology, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology (HUST), 430030, Wuhan, China.
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA.
- Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 200120, Shanghai, China.
| |
Collapse
|
109
|
Histone Demethylase KDM4C Stimulates the Proliferation of Prostate Cancer Cells via Activation of AKT and c-Myc. Cancers (Basel) 2019; 11:cancers11111785. [PMID: 31766290 PMCID: PMC6896035 DOI: 10.3390/cancers11111785] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 10/25/2019] [Accepted: 11/08/2019] [Indexed: 01/12/2023] Open
Abstract
Our three-dimensional organotypic culture revealed that human histone demethylase (KDM) 4C, a histone lysine demethylase, hindered the acini morphogenesis of RWPE-1 prostate cells, suggesting its potential oncogenic role. Knockdown (KD) of KDM4C suppressed cell proliferation, soft agar colony formation, and androgen receptor (AR) transcriptional activity in PCa cells as well as reduced tumor growth of human PCa cells in zebrafish xenotransplantation assay. Micro-Western array (MWA) analysis indicated that KD of KDM4C protein decreased the phosphorylation of AKT, c-Myc, AR, mTOR, PDK1, phospho-PDK1 S241, KDM8, and proteins involved in cell cycle regulators, while it increased the expression of PTEN. Fluorescent microscopy revealed that KDM4C co-localized with AR and c-Myc in the nuclei of PCa cells. Overexpression of either AKT or c-Myc rescued the suppressive effect of KDM4C KD on PCa cell proliferation. Echoing the above findings, the mRNA and protein expression of KDM4C was higher in human prostate tumor tissues as compared to adjacent normal prostate tissues, and higher KDM4C protein expression in prostate tumors correlated to higher protein expression level of AKT and c-Myc. In conclusion, KDM4C promotes the proliferation of PCa cells via activation of c-Myc and AKT.
Collapse
|
110
|
Ma X, Biswas A, Hammes SR. Paxillin regulated genomic networks in prostate cancer. Steroids 2019; 151:108463. [PMID: 31344408 PMCID: PMC6802295 DOI: 10.1016/j.steroids.2019.108463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 01/26/2023]
Abstract
Paxillin is extensively involved in focal adhesion signaling and kinase signaling throughout the plasma membrane and cytoplasm. However, recent studies in prostate cancer suggest that paxillin also plays a critical role in regulating gene expression within the nucleus, serving as a liaison between cytoplasmic and nuclear MAPK and Androgen Receptor (AR) signaling. Here we used RNA-seq to examine the paxillin-regulated transcriptome in several human prostate cancer cell lines. First, we examined paxillin effects on androgen-mediated transcription in control or paxillin-depleted AR-positive LNCaP and C4-2 human prostate cancer cells. In androgen-dependent LNCaP cells, we found over 1000 paxillin-dependent androgen-responsive genes, some of which are involved in endocrine therapy resistance. Most paxillin-dependent AR-mediated genes in LNCaP cells were no longer paxillin-dependent in androgen-sensitive, castration-resistant C4-2 cells, suggesting that castration-resistance may markedly alter paxillin effects on genomic AR signaling. To examine the paxillin-regulated transcriptome in the absence of androgen signaling, we performed RNA-seq in AR-negative PC3 human prostate cancer cells. Paxillin enhanced several pro-proliferative pathways, including the CyclinD/Rb/E2F and DNA replication/repair pathways. Additionally, paxillin suppressed pro-apoptotic genes, including CASP1 and TNFSF10. Quantitative PCR confirmed that these pathways are similarly regulated by paxillin in LNCaP and C4-2 cells. Functional studies showed that, while paxillin stimulated cell proliferation, it had minimum effect on apoptosis. Thus, paxillin appears to be an important transcriptional regulator in prostate cancer, and analysis of its transcriptome might lead to novel approaches toward the diagnosis and treatment of this important disease.
Collapse
Affiliation(s)
- Xiaoting Ma
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical School, Rochester, NY, United States
| | - Anindita Biswas
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, Lansing, MI, United States
| | - Stephen R Hammes
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical School, Rochester, NY, United States.
| |
Collapse
|
111
|
Mulholland EJ, Green WP, Buckley NE, McCarthy HO. Exploring the Potential of MicroRNA Let-7c as a Therapeutic for Prostate Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:927-937. [PMID: 31760377 PMCID: PMC6883330 DOI: 10.1016/j.omtn.2019.09.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 09/02/2019] [Accepted: 09/08/2019] [Indexed: 01/20/2023]
Abstract
Prostate cancer (PCa) is one of the leading causes of mortality worldwide and often presents with aberrant microRNA (miRNA) expression. Identifying and understanding the unique expression profiles could aid in the detection and treatment of this disease. This review aims to identify miRNAs as potential therapeutic targets for PCa. Three bio-informatic searches were conducted to identify miRNAs that are reportedly implicated in the pathogenesis of PCa. Only hsa-Lethal-7 (let-7c), recognized for its role in PCa pathogenesis, was common to all three databases. Three further database searches were conducted to identify known targets of hsa-let-7c. Four targets were identified, HMGA2, c-Myc (MYC), TRAIL, and CASP3. An extensive review of the literature was undertaken to assess the role of hsa-let-7c in the progression of other malignancies and to evaluate its potential as a therapeutic target for PCa. The heterogeneous nature of cancer makes it logical to develop mechanisms by which the treatment of malignancies is tailored to an individual, harnessing specific knowledge of the underlying biology of the disease. Resetting cellular miRNA levels is an exciting prospect that will allow this ambition to be realized.
Collapse
Affiliation(s)
- Eoghan J Mulholland
- Gastrointestinal Stem Cell Biology Laboratory, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - William P Green
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland
| | - Niamh E Buckley
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland.
| |
Collapse
|
112
|
Lin X, Kapoor A, Gu Y, Chow MJ, Xu H, Major P, Tang D. Assessment of biochemical recurrence of prostate cancer (Review). Int J Oncol 2019; 55:1194-1212. [PMID: 31638194 PMCID: PMC6831208 DOI: 10.3892/ijo.2019.4893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
The assessment of the risk of biochemical recurrence (BCR) is critical in the management of males with prostate cancer (PC). Over the past decades, a comprehensive effort has been focusing on improving risk stratification; a variety of models have been constructed using PC-associated pathological features and molecular alterations occurring at the genome, protein and RNA level. Alterations in RNA expression (lncRNA, miRNA and mRNA) constitute the largest proportion of the biomarkers of BCR. In this article, we systemically review RNA-based BCR biomarkers reported in PubMed according to the PRISMA guidelines. Individual miRNAs, mRNAs, lncRNAs and multi-gene panels, including the commercially available signatures, Oncotype DX and Prolaris, will be discussed; details related to cohort size, hazard ratio and 95% confidence intervals will be provided. Mechanistically, these individual biomarkers affect multiple pathways critical to tumorigenesis and progression, including epithelial-mesenchymal transition (EMT), phosphatase and tensin homolog (PTEN), Wnt, growth factor receptor, cell proliferation, immune checkpoints and others. This variety in the mechanisms involved not only validates their associations with BCR, but also highlights the need for the coverage of multiple pathways in order to effectively stratify the risk of BCR. Updates of novel biomarkers and their mechanistic insights are considered, which suggests new avenues to pursue in the prediction of BCR. Additionally, the management of patients with BCR and the potential utility of the stratification of the risk of BCR in salvage treatment decision making for these patients are briefly covered. Limitations will also be discussed.
Collapse
Affiliation(s)
- Xiaozeng Lin
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anil Kapoor
- The Research Institute of St. Joe's Hamilton, St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Yan Gu
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Mathilda Jing Chow
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Hui Xu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Pierre Major
- Division of Medical Oncology, Department of Oncology, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Damu Tang
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
113
|
Yu C, Hu K, Nguyen D, Wang ZA. From genomics to functions: preclinical mouse models for understanding oncogenic pathways in prostate cancer. Am J Cancer Res 2019; 9:2079-2102. [PMID: 31720076 PMCID: PMC6834478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/10/2019] [Indexed: 06/10/2023] Open
Abstract
Next-generation sequencing has revealed numerous genomic alterations that induce aberrant signaling activities in prostate cancer (PCa). Among them are pathways affecting multiple cancer types, including the PI3K/AKT/mTOR, p53, Rb, Ras/Raf/MAPK, Myc, FGF, and Wnt signaling pathways, as well as ones that are prominent in PCa, including alterations in genes of AR signaling, the ETS family, NKX3.1, and SPOP. Cross talk among the oncogenic pathways can confer PCa resistance to therapy, particularly in advanced tumors, which are castration-resistant or show neuroendocrine features. Various experimental models, such as cancer cell lines, animal models, and patient-derived xenografts and organoids have been utilized to dissect PCa progression mechanisms. Here, we review the current preclinical mouse models for studying the most commonly altered pathways in PCa, with an emphasis on their interplays. We highlight the power of genetically engineered mouse models (GEMMs) in translating genomic discoveries into understanding of the functions of these oncogenic events in vivo. Developing and analyzing PCa mouse models will undoubtedly continue to offer new insights into tumor biology and guide novel rationalized therapy.
Collapse
Affiliation(s)
- Chuan Yu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Kevin Hu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Daniel Nguyen
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Zhu A Wang
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| |
Collapse
|
114
|
Dong H, Hu J, Wang L, Qi M, Lu N, Tan X, Yang M, Bai X, Zhan X, Han B. SOX4 is activated by C-MYC in prostate cancer. Med Oncol 2019; 36:92. [PMID: 31560094 DOI: 10.1007/s12032-019-1317-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/12/2019] [Indexed: 11/30/2022]
Abstract
Although MYC proto-oncogene (C-MYC) amplification has been consistently reported to be a potential marker for prostate cancer (PCa) progression and prognosis, the clinicopathological and prognostic significance of C-MYC protein expression remains controversial. Overexpression of SOX4 has been shown to play important roles in multiple cancers including PCa. However, the link between these two critical genetic aberrations is unclear. In the current study, we showed that C-MYC was overexpressed in 16.2% (17/105) of Chinese patients with localized PCa. Overexpression of C-MYC was significantly associated with high Gleason scores (P = 0.012) and high Ki67 labeling index (P = 0.005). C-MYC overexpression was correlated with cancer-related mortality and suggested to be an unfavorable prognostic factor in Chinese PCa patients (P = 0.018). Overexpression of C-MYC is associated with SOX4 overexpression in PCa tissues. Notably, SOX4 is a direct target gene of C-MYC; C-MYC activates SOX4 expression via binding to its promoter. In addition, Co-IP analysis demonstrated a physical interaction between C-MYC and SOX4 protein in PCa cells. Clinically, C-MYC+/SOX4+ characterized poor prognosis in a subset of PCa patients. In total, C-MYC overexpression may contribute to PCa progression by activating SOX4. Our findings highlight an important role of C-MYC/SOX4 in PCa progression in a subset of PCa patients.
Collapse
Affiliation(s)
- Hongyan Dong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
- Department of Pathology, Linyi People's Hospital, Linyi, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Lin Wang
- Research Center for Medical Biotechnology, Shandong Academy of Medical Sciences, Jinan, China
| | - Mei Qi
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Ning Lu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Xiao Tan
- Department of Pathology, Linyi People's Hospital, Linyi, China
| | - Muyi Yang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Xinnuo Bai
- Department of Human Biology, University of Toronto, Toronto, M5S3J6, Canada
| | - Xuemei Zhan
- Department of Pathology, Linyi People's Hospital, Linyi, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
- Department of Pathology, Shandong University Qilu Hospital, Jinan, China.
| |
Collapse
|
115
|
Labbé DP, Zadra G, Yang M, Reyes JM, Lin CY, Cacciatore S, Ebot EM, Creech AL, Giunchi F, Fiorentino M, Elfandy H, Syamala S, Karoly ED, Alshalalfa M, Erho N, Ross A, Schaeffer EM, Gibb EA, Takhar M, Den RB, Lehrer J, Karnes RJ, Freedland SJ, Davicioni E, Spratt DE, Ellis L, Jaffe JD, DʼAmico AV, Kantoff PW, Bradner JE, Mucci LA, Chavarro JE, Loda M, Brown M. High-fat diet fuels prostate cancer progression by rewiring the metabolome and amplifying the MYC program. Nat Commun 2019; 10:4358. [PMID: 31554818 PMCID: PMC6761092 DOI: 10.1038/s41467-019-12298-z] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Systemic metabolic alterations associated with increased consumption of saturated fat and obesity are linked with increased risk of prostate cancer progression and mortality, but the molecular underpinnings of this association are poorly understood. Here, we demonstrate in a murine prostate cancer model, that high-fat diet (HFD) enhances the MYC transcriptional program through metabolic alterations that favour histone H4K20 hypomethylation at the promoter regions of MYC regulated genes, leading to increased cellular proliferation and tumour burden. Saturated fat intake (SFI) is also associated with an enhanced MYC transcriptional signature in prostate cancer patients. The SFI-induced MYC signature independently predicts prostate cancer progression and death. Finally, switching from a high-fat to a low-fat diet, attenuates the MYC transcriptional program in mice. Our findings suggest that in primary prostate cancer, dietary SFI contributes to tumour progression by mimicking MYC over expression, setting the stage for therapeutic approaches involving changes to the diet.
Collapse
Affiliation(s)
- David P Labbé
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Urology, Department of Surgery, McGill University and Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Giorgia Zadra
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Meng Yang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jaime M Reyes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Charles Y Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Stefano Cacciatore
- Cancer Genomics Group, International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Ericka M Ebot
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Amanda L Creech
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Francesca Giunchi
- Pathology Service, Addarii Institute of Oncology, S-Orsola-Malpighi Hospital, Bologna, IT, Italy
| | - Michelangelo Fiorentino
- Pathology Service, Addarii Institute of Oncology, S-Orsola-Malpighi Hospital, Bologna, IT, Italy
| | - Habiba Elfandy
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sudeepa Syamala
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Ashley Ross
- James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | - Robert B Den
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | | | - R Jeffrey Karnes
- Department of Urology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Stephen J Freedland
- Department of Surgery, Division of Urology, Center for Integrated Research on Cancer and Lifestyle, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Surgery Section, Durham Veteran Affairs Medical Center, Durham, NC, USA
| | | | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Leigh Ellis
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jacob D Jaffe
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Anthony V DʼAmico
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip W Kantoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jorge E Chavarro
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA.
- Department of Pathology and Laboratory Medicine, Weil Cornell Medicine, New York Presbyterian-Weill Cornell Campus, New York, NY, USA.
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
116
|
Umbreen S, Banday MM, Jamroze A, Mansini AP, Ganaie AA, Ferrari MG, Maqbool R, Beigh FH, Murugan P, Morrissey C, Corey E, Konety BR, Saleem M. COMMD3:BMI1 Fusion and COMMD3 Protein Regulate C-MYC Transcription: Novel Therapeutic Target for Metastatic Prostate Cancer. Mol Cancer Ther 2019; 18:2111-2123. [PMID: 31467179 DOI: 10.1158/1535-7163.mct-19-0150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/24/2019] [Accepted: 08/20/2019] [Indexed: 11/16/2022]
Abstract
Gene rearrangement is reported to be associated to the aggressive phenotype and poor prognosis in prostate cancer. We identified a gene fusion between a transcription repressor (BMI1) and transcriptional factor (COMMD3) in human prostate cancer. We show that COMMD3:BMI1 fusion expression is significantly increased in prostate cancer disease in an order: normal tissue < primary < metastatic tumors (Mets). Although elevated TMPRSS-ERG/ETV fusion is reported in prostate cancer, we identified a subtype of Mets exhibiting low TMPRSS:ETV and high COMMD3:BMI1 We delineated the mechanism and function of COMMD3 and COMMD3:BMI1 in prostate cancer. We show that COMMD3 level is elevated in prostate cancer cell models, PDX models (adenocarcinoma, NECaP), and Mets. The analysis of TCGA/NIH/GEO clinical data showed a positive correlation between increased COMMD3 expression to the disease recurrence and poor survival in prostate cancer. We show that COMMD3 drives proliferation of normal cells and promotes migration/invasiveness of neoplastic cells. We show that COMMD3:BMI1 and COMMD3 regulate C-MYC transcription and C-MYC downstream pathway. The ChIP analysis showed that COMMD3 protein is recruited at the promoter of C-MYC gene. On the basis of these data, we investigated the relevance of COMMD3:BMI1 and COMMD3 as therapeutic targets using in vitro and xenograft mouse models. We show that siRNA-mediated targeting of COMMD3:BMI1 and COMMD3 significantly decreases (i) C-MYC expression in BRD/BET inhibitor-resistant cells, (ii) proliferation/invasion in vitro, and (iii) growth of prostate cancer cell tumors in mice. The IHC analysis of tumors confirmed the targeting of COMMD3-regulated molecular pathway under in vivo conditions. We conclude that COMMD3:BMI1 and COMMD3 are potential progression biomarkers and therapeutic targets of metastatic prostate cancer.
Collapse
Affiliation(s)
- Syed Umbreen
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Queens University, Belfast, Northern Ireland
| | - Mudassir Meraj Banday
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Anmbreen Jamroze
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Hormel Institute, Austin, Minnesota
| | - Adrian P Mansini
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Arsheed A Ganaie
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Marina G Ferrari
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Raihana Maqbool
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Firdous H Beigh
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | | | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Badrinath R Konety
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Mohammad Saleem
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
117
|
IGHG1 Regulates Prostate Cancer Growth via the MEK/ERK/c-Myc Pathway. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7201562. [PMID: 31355278 PMCID: PMC6637713 DOI: 10.1155/2019/7201562] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 12/27/2022]
Abstract
Increasing evidence indicates that immunoglobulins are important for the regulation of various cancers including prostate cancer (PCa). However, the underlying mechanisms of IgG regulated PCa development remain to be further explored. Here, we demonstrated that IgG1 heavy chain (IGHG1) was increased in tissues from PCa patients. Inhibition of IGHG1 by antibody blocking or genetic knockdown suppressed cell growth and induced cell cycle arrest and ultimate apoptosis. Expression levels of c-Myc were positively correlated with the levels of IGHG1. Furthermore, MEK/ERK/c-Myc pathway lied downstream of IGHG1 in cultured prostate cancer cells. Inhibition of IGHG1 restrained the tumor growth in nude mice and inactivated MEK/ERK/c-Myc pathway both in vitro and in vivo. These findings suggest that IGHG1 play a crucial role during the development of prostate cancer and inhibition of IGHG1 may be a potential therapy in the treatment of PCa.
Collapse
|
118
|
Islam MT, Zhou X, Chen F, Khan MA, Fu J, Chen H. Targeting the signalling pathways regulated by deubiquitinases for prostate cancer therapeutics. Cell Biochem Funct 2019; 37:304-319. [PMID: 31062387 DOI: 10.1002/cbf.3401] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/06/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022]
Abstract
Prostate cancer (PCa) is the most common cancer diagnosed and the second most common cause of cancer-related death in men worldwide. The current androgen deprivation therapy for PCa cannot fully cure this disease. Moreover, androgen receptor gene amplification and mutation are associated with PCa to develop castration-resistant prostate cancer (CRPC). This review focuses on the deubiquitinases (DUBs) involved in PCa development and progression. For PCa development and progression, several cellular pathways are regulated by specific DUBs which are also highlighted in here. The ubiquitin-specific proteases (USPs), a family member of DUBs mostly involved in the regulation of cellular pathways for PCa development, and the ubiquitin C-terminal hydrolases (UCHs), another family member of DUBs, are responsible for PCa metastasis. Small molecular inhibitors against DUBs can inhibit or reduce the level of specific DUBs through the regulation of cellular pathway to treat this disease. Some small molecular inhibitors are already identified against some of the DUBs, but very few of them are clinically proved in PCa. So, to find out other DUBs involving in the regulation of PCa-related pathways and to develop more effective small molecule inhibitors with greater potency would be a great idea to target PCa cells for future therapeutics and drug development with or without the combination of other anticancer drugs. SIGNIFICANCE OF THE STUDY: This review is targeting DUB proteins which are responsible for PCa induction, proliferation, and metastasis by highlighting their signalling pathway so that the readers can get information about other mechanisms for PCa besides androgen receptor pathway and helps to find other oncogenic DUBs involving in these signalling pathways. This review also hopes to find other oncogenic DUBs involving in PCa-related signalling pathways or to find the DUBs that can regulate multiple oncogenic signalling pathways which might be a good target for PCa therapeutics. In addition, there are some small molecule inhibitors that can inhibit the oncogenic DUBs and thus able to control the oncogenic pathways which would be a novel strategy to treat CRPC by using DUB inhibitor combined with or without other anticancer drugs.
Collapse
Affiliation(s)
- Md Tariqul Islam
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| | - Xi Zhou
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| | - Fangzhi Chen
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Md Asaduzzaman Khan
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Hanchun Chen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
119
|
Berger A, Brady NJ, Bareja R, Robinson B, Conteduca V, Augello MA, Puca L, Ahmed A, Dardenne E, Lu X, Hwang I, Bagadion AM, Sboner A, Elemento O, Paik J, Yu J, Barbieri CE, Dephoure N, Beltran H, Rickman DS. N-Myc-mediated epigenetic reprogramming drives lineage plasticity in advanced prostate cancer. J Clin Invest 2019; 129:3924-3940. [PMID: 31260412 DOI: 10.1172/jci127961] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Despite recent therapeutic advances, prostate cancer remains a leading cause of cancer-related death. A subset of castration resistant prostate cancers become androgen receptor (AR) signaling-independent and develop neuroendocrine prostate cancer (NEPC) features through lineage plasticity. These NEPC tumors, associated with aggressive disease and poor prognosis, are driven, in part, by aberrant expression of N-Myc, through mechanisms that remain unclear. Integrative analysis of the N-Myc transcriptome, cistrome and interactome using in vivo, in vitro and ex vivo models (including patient-derived organoids) identified a lineage switch towards a neural identity associated with epigenetic reprogramming. N-Myc and known AR-co-factors (e.g., FOXA1 and HOXB13) overlapped, independently of AR, at genomic loci implicated in neural lineage specification. Moreover, histone marks specifically associated with lineage-defining genes were reprogrammed by N-Myc. We also demonstrated that the N-Myc-induced molecular program accurately classifies our cohort of patients with advanced prostate cancer. Finally, we revealed the potential for EZH2 inhibition to reverse the N-Myc-induced suppression of epithelial lineage genes. Altogether, our data provide insights on how N-Myc regulates lineage plasticity and epigenetic reprogramming associated with lineage-specification. The N-Myc signature we defined could also help predict the evolution of prostate cancer and thus better guide the choice of future therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Rohan Bareja
- Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital
| | - Brian Robinson
- Department of Pathology and Laboratory Medicine.,Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital
| | | | | | | | - Adnan Ahmed
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | | | - Xiaodong Lu
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Inah Hwang
- Department of Pathology and Laboratory Medicine
| | | | - Andrea Sboner
- Department of Pathology and Laboratory Medicine.,Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Department of Physiology and Biophysics, Institute for Computational Biomedicine, and.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Department of Physiology and Biophysics, Institute for Computational Biomedicine, and.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Jindan Yu
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Christopher E Barbieri
- Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Department of Urology, and.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Noah Dephoure
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Himisha Beltran
- Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Department of Medicine.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - David S Rickman
- Department of Pathology and Laboratory Medicine.,Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
120
|
Kosalai ST, Morsy MHA, Papakonstantinou N, Mansouri L, Stavroyianni N, Kanduri C, Stamatopoulos K, Rosenquist R, Kanduri M. EZH2 upregulates the PI3K/AKT pathway through IGF1R and MYC in clinically aggressive chronic lymphocytic leukaemia. Epigenetics 2019; 14:1125-1140. [PMID: 31216925 PMCID: PMC6773411 DOI: 10.1080/15592294.2019.1633867] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
EZH2 is overexpressed in poor-prognostic chronic lymphocytic leukaemia (CLL) cases, acting as an oncogene; however, thus far, the EZH2 target genes in CLL have not been disclosed. In this study, using ChIP-sequencing, we identified EZH2 and H3K27me3 target genes in two prognostic subgroups of CLL with distinct prognosis and outcome, i.e., cases with unmutated (U-CLL, n = 6) or mutated IGHV genes (M-CLL, n = 6). While the majority of oncogenic pathways were equally enriched for EZH2 target genes in both prognostic subgroups, PI3K pathway genes were differentially bound by EZH2 in U-CLL versus M-CLL. The occupancy of EZH2 for selected PI3K pathway target genes was validated in additional CLL samples (n = 16) and CLL cell lines using siRNA-mediated EZH2 downregulation and ChIP assays. Intriguingly, we found that EZH2 directly binds to the IGF1R promoter along with MYC and upregulates IGF1R expression in U-CLL, leading to downstream PI3K activation. By investigating an independent CLL cohort (n = 96), a positive correlation was observed between EZH2 and IGF1R expression with higher levels in U-CLL compared to M-CLL. Accordingly, siRNA-mediated downregulation of either EZH2, MYC or IGF1R and treatment with EZH2 and MYC pharmacological inhibitors in the HG3 CLL cell line induced a significant reduction in PI3K pathway activation. In conclusion, we characterize for the first time EZH2 target genes in CLL revealing a hitherto unknown implication of EZH2 in modulating the PI3K pathway in a non-canonical, PRC2-independent way, with potential therapeutic implications considering that PI3K inhibitors are effective therapeutic agents for CLL.
Collapse
Affiliation(s)
- Subazini Thankaswamy Kosalai
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | | | - Nikos Papakonstantinou
- Institute of Applied Biosciences, Center for Research and Technology Hellas , Thessaloniki , Greece
| | - Larry Mansouri
- Department of Molecular Medicine and Surgery, Karolinska Institutet , Stockholm , Sweden
| | - Niki Stavroyianni
- Hematology Department and HCT Unit, G. Papanicolaou Hospital , Thessaloniki , Greece
| | - Chandrasekhar Kanduri
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Kostas Stamatopoulos
- Institute of Applied Biosciences, Center for Research and Technology Hellas , Thessaloniki , Greece
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet , Stockholm , Sweden
| | - Meena Kanduri
- Department of Clinical chemistry and Transfusion medicine, Sahlgrenska University Hospital , Gothenburg , Sweden
| |
Collapse
|
121
|
[Prostate pathology recommendations from the Uropathology working group of the Spanish Society of Pathology]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2019; 52:167-177. [PMID: 31213258 DOI: 10.1016/j.patol.2019.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 11/24/2022]
Abstract
These guidelines from the uropathology working group of the Spanish Society of Pathology (SEAP) are based on the European and ISUP 2015 recommendations and those of the College of American Pathologists, as well as the latest WHO 2016, TNM (AJCC) 2017 classifications. They include recommendations for specimen sampling, macro- and microscopic examination and immunohistochemistry. Gleason patterns are specified: Gleason pattern 3 includes hyperplastic, atrophic and microcystic glands, while pattern 4 includes all cribriform or glomeruloid glands. The Gleason score in prostatectomy specimens may change; if a tertiary pattern occurs in more than 5% of the tumour, it becomes a secondary pattern. In both biopsies and prostatectomy specimens, if the Gleason score is 7, the percentage of pattern 4 should be stated. Gleason scoring in tumor variants and special situations should also be specified. These recommendations should be adapted according to the resources available.
Collapse
|
122
|
Paglia S, Sollazzo M, Di Giacomo S, Strocchi S, Grifoni D. Exploring MYC relevance to cancer biology from the perspective of cell competition. Semin Cancer Biol 2019; 63:49-59. [PMID: 31102666 DOI: 10.1016/j.semcancer.2019.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022]
Abstract
Cancer has long been regarded and treated as a foreign body appearing by mistake inside a living organism. However, now we know that cancer cells communicate with neighbours, thereby creating modified environments able to support their unusual need for nutrients and space. Understanding the molecular basis of these bi-directional interactions is thus mandatory to approach the complex nature of cancer. Since their discovery, MYC proteins have been showing to regulate a steadily increasing number of processes impacting cell fitness, and are consistently found upregulated in almost all human tumours. Of interest, MYC takes part in cell competition, an evolutionarily conserved fitness comparison strategy aimed at detecting weakened cells, which are then committed to death, removed from the tissue and replaced by fitter neighbours. During physiological development, MYC-mediated cell competition is engaged to eliminate cells with suboptimal MYC levels, so as to guarantee selective growth of the fittest and proper homeostasis, while transformed cells expressing high levels of MYC coopt cell competition to subvert tissue constraints, ultimately disrupting homeostasis. Therefore, the interplay between cells with different MYC levels may result in opposite functional outcomes, depending on the nature of the players. In the present review, we describe the most recent findings on the role of MYC-mediated cell competition in different contexts, with a special emphasis on its impact on cancer initiation and progression. We also discuss the relevance of competition-associated cell death to cancer disease.
Collapse
Affiliation(s)
- Simona Paglia
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| | - Manuela Sollazzo
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| | - Simone Di Giacomo
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| | - Silvia Strocchi
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| | - Daniela Grifoni
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| |
Collapse
|
123
|
Gao Y, Wei L, Wang C, Huang Y, Li W, Li T, Mo C, Qin H, Zhong X, Wang Y, Tan A, Mo Z, Jiang Y, Hu Y. Chronic prostatitis alters the prostatic microenvironment and accelerates preneoplastic lesions in C57BL/6 mice. Biol Res 2019; 52:30. [PMID: 31088536 PMCID: PMC6518623 DOI: 10.1186/s40659-019-0237-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 04/29/2019] [Indexed: 12/24/2022] Open
Abstract
Background Chronic prostatitis has been supposed to be associated with preneoplastic lesions and cancer development. The objective of this study was to examine how chronic inflammation results in a prostatic microenvironment and gene mutation in C57BL/6 mice. Methods Immune and bacterial prostatitis mouse models were created through abdominal subcutaneous injection of rat prostate extract protein immunization (EAP group) or transurethral instillation of uropathogenic E. coli 1677 (E. coli group). Prostate histology, serum cytokine level, and genome-wide exome (GWE) sequences were examined 1, 3, and 6 months after immunization or injection. Result In the EAP and E. coli groups, immune cell infiltrations were observed in the first and last months of the entire experiment. After 3 months, obvious proliferative inflammatory atrophy (PIA) and prostatic intraepithelial neoplasia (PIN) were observed accompanied with fibrosis hyperplasia in stroma. The decrease in basal cells (Cytokeratin (CK) 5+/p63+) and the accumulation of luminal epithelial cells (CK8+) in the PIA or PIN area indicated that the basal cells were damaged or transformed into different luminal cells. Hic1, Zfp148, and Mfge8 gene mutations were detected in chronic prostatitis somatic cells. Conclusion Chronic prostatitis induced by prostate extract protein immunization or E. coli infection caused a reactive prostatic inflammation microenvironment and resulted in tissue damage, aberrant atrophy, hyperplasia, and somatic genome mutation. Electronic supplementary material The online version of this article (10.1186/s40659-019-0237-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yong Gao
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lijuan Wei
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chenbang Wang
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuanjie Huang
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Weidong Li
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Tianyu Li
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chaohua Mo
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huali Qin
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoge Zhong
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yun Wang
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Aihua Tan
- Department of Chemotherapy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.,Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, 530021, Guangxi, China
| | - Yonghua Jiang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China. .,Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, 530021, Guangxi, China. .,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, 530021, Guangxi, China.
| | - Yanling Hu
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China. .,Department of Chemotherapy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China. .,Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, 530021, Guangxi, China. .,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, 530021, Guangxi, China.
| |
Collapse
|
124
|
Elliott B, Millena AC, Matyunina L, Zhang M, Zou J, Wang G, Zhang Q, Bowen N, Eaton V, Webb G, Thompson S, McDonald J, Khan S. Essential role of JunD in cell proliferation is mediated via MYC signaling in prostate cancer cells. Cancer Lett 2019; 448:155-167. [PMID: 30763715 PMCID: PMC6414252 DOI: 10.1016/j.canlet.2019.02.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/12/2022]
Abstract
JunD, a member of the AP-1 family, is essential for cell proliferation in prostate cancer (PCa) cells. We recently demonstrated that JunD knock-down (KD) in PCa cells results in cell cycle arrest in G1-phase concomitant with a decrease in cyclin D1, Ki67, and c-MYC, but an increase in p21 levels. Furthermore, the over-expression of JunD significantly increased proliferation suggesting JunD regulation of genes required for cell cycle progression. Here, employing gene expression profiling, quantitative proteomics, and validation approaches, we demonstrate that JunD KD is associated with distinct gene and protein expression patterns. Comparative integrative analysis by Ingenuity Pathway Analysis (IPA) identified 1) cell cycle control/regulation as the top canonical pathway whose members exhibited a significant decrease in their expression following JunD KD including PRDX3, PEA15, KIF2C, and CDK2, and 2) JunD dependent genes are associated with cell proliferation, with MYC as the critical downstream regulator. Conversely, JunD over-expression induced the expression of the above genes including c-MYC. We conclude that JunD is a crucial regulator of cell cycle progression and inhibiting its target genes may be an effective approach to block prostate carcinogenesis.
Collapse
Affiliation(s)
- Bethtrice Elliott
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Ana Cecilia Millena
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Lilya Matyunina
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30309, USA
| | - Mengnan Zhang
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30309, USA
| | - Jin Zou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Guangdi Wang
- Department of Chemistry, RCMI Cancer Research Center, Xavier University, 1 Drexel Drive, New Orleans, LA, 70125, USA
| | - Qiang Zhang
- Department of Chemistry, RCMI Cancer Research Center, Xavier University, 1 Drexel Drive, New Orleans, LA, 70125, USA
| | - Nathan Bowen
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Vanessa Eaton
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Gabrielle Webb
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - Shadyra Thompson
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA
| | - John McDonald
- Integrated Cancer Research Center, School of Biological Sciences, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30309, USA
| | - Shafiq Khan
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr. SW, Atlanta, GA, 30314, USA.
| |
Collapse
|
125
|
Makwana V, Ryan P, Patel B, Dukie SA, Rudrawar S. Essential role of O-GlcNAcylation in stabilization of oncogenic factors. Biochim Biophys Acta Gen Subj 2019; 1863:1302-1317. [PMID: 31034911 DOI: 10.1016/j.bbagen.2019.04.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
A reversible post-translational protein modification which involves addition of N-acetylglucosamine (GlcNAc) onto hydroxyl groups of serine and/or threonine residues which is known as O-GlcNAcylation, has emerged as a potent competitor of phosphorylation. This glycosyltransfer reaction is catalyzed by the enzyme O-linked β-N-acetylglucosamine transferase (OGT). This enzyme uses uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), the end product of hexosamine biosynthetic pathway, to modify numerous nuclear and cytosolic proteins. O-GlcNAcylation influences cancer cell metabolism in such a way that hyper-O-GlcNAcylation is considered as a prominent trait of many cancers, and is proposed as a major factor enabling cancer cell proliferation and progression. Growing evidence supports a connection between O-GlcNAcylation and major oncogenic factors, including for example, c-MYC, HIF-1α, and NF-κB. A comprehensive study of the roles of O-GlcNAc modification of oncogenic factors is warranted as a thorough understanding may help drive advances in cancer diagnosis and therapy. The focus of this article is to highlight the interplay between oncogenic factors and O-GlcNAcylation along with OGT in cancer cell proliferation and survival. The prospects for OGT inhibitors will also be discussed.
Collapse
Affiliation(s)
- Vivek Makwana
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Philip Ryan
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Bhautikkumar Patel
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Shailendra-Anoopkumar Dukie
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast 4222, Australia.
| | - Santosh Rudrawar
- School of Pharmacy and Pharmacology, Griffith University, Gold Coast, Queensland 4222, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland 4222, Australia; Quality Use of Medicines Network, Griffith University, Gold Coast 4222, Australia.
| |
Collapse
|
126
|
Salem O, Hansen CG. The Hippo Pathway in Prostate Cancer. Cells 2019; 8:E370. [PMID: 31018586 PMCID: PMC6523349 DOI: 10.3390/cells8040370] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 02/07/2023] Open
Abstract
Despite recent efforts, prostate cancer (PCa) remains one of the most common cancers in men. Currently, there is no effective treatment for castration-resistant prostate cancer (CRPC). There is, therefore, an urgent need to identify new therapeutic targets. The Hippo pathway and its downstream effectors-the transcriptional co-activators, Yes-associated protein (YAP) and its paralog, transcriptional co-activator with PDZ-binding motif (TAZ)-are foremost regulators of stem cells and cancer biology. Defective Hippo pathway signaling and YAP/TAZ hyperactivation are common across various cancers. Here, we draw on insights learned from other types of cancers and review the latest advances linking the Hippo pathway and YAP/TAZ to PCa onset and progression. We examine the regulatory interaction between Hippo-YAP/TAZ and the androgen receptor (AR), as main regulators of PCa development, and how uncontrolled expression of YAP/TAZ drives castration resistance by inducing cellular stemness. Finally, we survey the potential therapeutic targeting of the Hippo pathway and YAP/TAZ to overcome PCa.
Collapse
Affiliation(s)
- Omar Salem
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| | - Carsten G Hansen
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| |
Collapse
|
127
|
Trabzonlu L, Kulac I, Zheng Q, Hicks JL, Haffner MC, Nelson WG, Sfanos KS, Ertunc O, Lotan TL, Heaphy CM, Meeker AK, Yegnasubramanian S, De Marzo AM. Molecular Pathology of High-Grade Prostatic Intraepithelial Neoplasia: Challenges and Opportunities. Cold Spring Harb Perspect Med 2019; 9:a030403. [PMID: 30082453 PMCID: PMC6444695 DOI: 10.1101/cshperspect.a030403] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A better understanding of the early stages of prostate cancer initiation, potentially arising from precursor lesions, may fuel development of powerful approaches for prostate cancer prevention or interception. The best-known candidate for such a precursor lesion has been referred to as high-grade prostatic intraepithelial neoplasia (HGPIN). Although there is significant evidence supporting the notion that such HGPIN lesions can give rise to invasive adenocarcinomas of the prostate, there are also numerous complicating considerations and evidence that cloud the picture in many instances. Notably, recent evidence has suggested that some fraction of such lesions that are morphologically consistent with HGPIN may actually be invasive carcinomas masquerading as HGPIN-a state that we term "postinvasive intraepithelial carcinoma" (PIC). Although the prevalence of such PIC lesions is not fully understood, this and other factors can confound the potential of identifying prostate precursors that can be targeted for disease prevention, interception, or treatment. Here, we review our current understanding of the morphological and molecular pathological features of prostate cancer precursor lesions.
Collapse
Affiliation(s)
- Levent Trabzonlu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Ibrahim Kulac
- Department of Pathology, Koc University School of Medicine, Istanbul 34010, Turkey
| | - Qizhi Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Jessica L Hicks
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Michael C Haffner
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - William G Nelson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
- The Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
- The Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Onur Ertunc
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - Christopher M Heaphy
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
- The Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Alan K Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
- The Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
- The Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
- The Brady Urological Research Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| |
Collapse
|
128
|
Zhao M, Qi M, Li X, Hu J, Zhang J, Jiao M, Bai X, Peng X, Han B. CUL4B/miR-33b/C-MYC axis promotes prostate cancer progression. Prostate 2019; 79:480-488. [PMID: 30609075 DOI: 10.1002/pros.23754] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/27/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Cullin 4B (CUL4B), a scaffold protein that assembles CRL4B ubiquitin ligase complexes, is overexpressed in many types of solid tumors and contributes to epigenetic silencing of tumor suppressors. However, its clinical significance and underlying molecular mechanisms in prostate cancer (PCa) remain unknown. METHODS The clinical significance of CUL4B in PCa was characterized by in silico method. RT-qPCR and Western blot were used to study the transcript and protein expression levels of CUL4B and C-MYC. Bioinformatics tools, chromatin immunoprecipitation (ChIP) and luciferase reporter assay were utilized to identify and characterize the microRNAs (miRNAs) regulated by CUL4B. The biological function of CUL4B and miR-33b-5p was evaluated by MTS, transwell, and wound healing assays, accordingly. RESULTS CUL4B is significantly overexpressed in PCa tissues compared with benign prostatic tissues and its overexpression is correlated with poor prognosis. CUL4B promotes proliferation and aggressiveness of PCa cells in vitro. Mechanistically, we demonstrate that CUL4B upregulates the expression of C-MYC at post-transcriptional level through epigenetic silencing of miR-33b-5p. Importantly, CUL4B-induced oncogenic activity in PCa by targeting C-MYC is repressed by miR-33b-5p. CONCLUSIONS Our results suggested a novel CUL4B/miR-33b/C-MYC axis implicated in PCa cell growth and progression. This might provide novel insight into how CUL4B contributed to PCa aggressiveness and progression.
Collapse
Affiliation(s)
- Mingfeng Zhao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
- Department of Pathology, Binzhou Medical University, Binzhou, China
| | - Mei Qi
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
| | - Xinjun Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
- Department of Pathology, Binzhou People's Hospital, Binzhou, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
| | - Jing Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Meng Jiao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
| | - Xinnuo Bai
- Department of Public Health Sciences, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Xijia Peng
- Human Biology Program, University of Toronto, Toronto, Ontario, Canada
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University QiLu Medical College, School of Basic Medical Sciences, Jinan, China
- Department of Pathology, Shandong University QiLu Hospital, Jinan, China
| |
Collapse
|
129
|
Braadland PR, Urbanucci A. Chromatin reprogramming as an adaptation mechanism in advanced prostate cancer. Endocr Relat Cancer 2019; 26:R211-R235. [PMID: 30844748 DOI: 10.1530/erc-18-0579] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
Tumor evolution is based on the ability to constantly mutate and activate different pathways under the selective pressure of targeted therapies. Epigenetic alterations including those of the chromatin structure are associated with tumor initiation, progression and drug resistance. Many cancers, including prostate cancer, present enlarged nuclei, and chromatin appears altered and irregular. These phenotypic changes are likely to result from epigenetic dysregulation. High-throughput sequencing applied to bulk samples and now to single cells has made it possible to study these processes in unprecedented detail. It is therefore timely to review the impact of chromatin relaxation and increased DNA accessibility on prostate cancer growth and drug resistance, and their effects on gene expression. In particular, we focus on the contribution of chromatin-associated proteins such as the bromodomain-containing proteins to chromatin relaxation. We discuss the consequence of this for androgen receptor transcriptional activity and briefly summarize wider gain-of-function effects on other oncogenic transcription factors and implications for more effective prostate cancer treatment.
Collapse
Affiliation(s)
- Peder Rustøen Braadland
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alfonso Urbanucci
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, Oslo, Norway
| |
Collapse
|
130
|
Mallappa S, Neeli PK, Karnewar S, Kotamraju S. Doxorubicin induces prostate cancer drug resistance by upregulation of ABCG4 through GSH depletion and CREB activation: Relevance of statins in chemosensitization. Mol Carcinog 2019; 58:1118-1133. [DOI: 10.1002/mc.22996] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/02/2019] [Accepted: 02/11/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Sreevidya Mallappa
- Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology; Hyderabad India
- Academy of Scientific and Innovative Research, Training and Development Complex; Chennai India
| | - Praveen K. Neeli
- Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology; Hyderabad India
- Academy of Scientific and Innovative Research, Training and Development Complex; Chennai India
| | - Santosh Karnewar
- Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology; Hyderabad India
- Academy of Scientific and Innovative Research, Training and Development Complex; Chennai India
| | - Srigiridhar Kotamraju
- Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology; Hyderabad India
- Academy of Scientific and Innovative Research, Training and Development Complex; Chennai India
| |
Collapse
|
131
|
Kido T, Li Y, Tanaka Y, Dahiya R, Chris Lau YF. The X-linked tumor suppressor TSPX downregulates cancer-drivers/oncogenes in prostate cancer in a C-terminal acidic domain dependent manner. Oncotarget 2019; 10:1491-1506. [PMID: 30863497 PMCID: PMC6407674 DOI: 10.18632/oncotarget.26673] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 01/31/2019] [Indexed: 01/02/2023] Open
Abstract
TSPX is a tumor suppressor gene located at Xp11.22, a prostate cancer susceptibility locus. It is ubiquitously expressed in most tissues but frequently downregulated in various cancers, including lung, brain, liver and prostate cancers. The C-terminal acidic domain (CAD) of TSPX is crucial for the tumor suppressor functions, such as inhibition of cyclin B/CDK1 phosphorylation and androgen receptor transactivation. Currently, the exact role of the TSPX CAD in transcriptional regulation of downstream genes is still uncertain. Using different variants of TSPX, we showed that overexpression of either TSPX, that harbors a CAD, or a CAD-truncated variant (TSPX[∆C]) drastically retarded cell proliferation in a prostate cancer cell line LNCaP, but cell death was induced only by overexpression of TSPX. Transcriptome analyses showed that TSPX or TSPX[∆C] overexpression downregulated multiple cancer-drivers/oncogenes, including MYC and MYB, in a CAD-dependent manner and upregulated various tumor suppressors in a CAD-independent manner. Datamining of transcriptomes of prostate cancer specimens in the Cancer Genome Atlas (TCGA) dataset confirmed the negative correlation between the expression level of TSPX and those of MYC and MYB in clinical prostate cancer, thereby supporting the hypothesis that the CAD of TSPX plays an important role in suppression of cancer-drivers/oncogenes in prostatic oncogenesis.
Collapse
Affiliation(s)
- Tatsuo Kido
- Division of Cell and Developmental Genetics, Department of Medicine, Veterans Affairs Medical Center, San Francisco, California, USA
- Institute for Human Genetics, University of California, San Francisco, California, USA
| | - Yunmin Li
- Division of Cell and Developmental Genetics, Department of Medicine, Veterans Affairs Medical Center, San Francisco, California, USA
- Institute for Human Genetics, University of California, San Francisco, California, USA
| | - Yuichiro Tanaka
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California, USA
| | - Rajvir Dahiya
- Department of Urology, Veterans Affairs Medical Center, San Francisco and University of California San Francisco, San Francisco, California, USA
| | - Yun-Fai Chris Lau
- Division of Cell and Developmental Genetics, Department of Medicine, Veterans Affairs Medical Center, San Francisco, California, USA
- Institute for Human Genetics, University of California, San Francisco, California, USA
| |
Collapse
|
132
|
Yang J, Guo Y, Lu C, Zhang R, Wang Y, Luo L, Zhang Y, Chu CH, Wang KJ, Obbad S, Yan W, Li X. Inhibition of Karyopherin beta 1 suppresses prostate cancer growth. Oncogene 2019; 38:4700-4714. [PMID: 30742095 PMCID: PMC6565446 DOI: 10.1038/s41388-019-0745-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 12/10/2018] [Accepted: 01/26/2019] [Indexed: 12/19/2022]
Abstract
Prostate cancer (PCa) initiation and progression requires activation of numerous oncogenic signaling pathways. Nuclear-cytoplasmic transport of oncogenic factors is mediated by Karyopherin proteins during cell transformation. However, the role of nuclear transporter proteins in PCa progression has not been well defined. Here, we report that the KPNB1, a key member of Karyopherin beta subunits, is highly expressed in advanced prostate cancers. Further study showed that targeting KPNB1 suppressed the proliferation of prostate cancer cells. The knockdown of KPNB1 reduced nuclear translocation of c-Myc, the expression of downstream cell cycle modulators, and phosphorylation of regulator of chromatin condensation 1 (RCC1), a key protein for spindle assembly during mitosis. Meanwhile, CHIP assay demonstrated the binding of c-Myc to KPNB1 promoter region, which indicated a positive feedback regulation of KPNB1 expression mediated by the c-Myc. In addition, NF-κB subunit p50 translocation to nuclei was blocked by KPNB1 inhibition, which led to an increase in apoptosis and a decrease in tumor sphere formation of PCa cells. Furthermore, subcutaneous xenograft tumor models with a stable knockdown of KPNB1 in C42B PCa cells validated that the inhibition of KPNB1 could suppress the growth of prostate tumor in vivo. Moreover, the intravenously administration of importazole, a specific inhibitor for KPNB1, effectively reduced PCa tumor size and weight in mice inoculated with PC3 PCa cells. In summary, our data established the functional link between KPNB1 and PCa prone c-Myc, NF-kB, and cell cycle modulators. More importantly, inhibition of KPNB1 could be a new therapeutic target for PCa treatment.
Collapse
Affiliation(s)
- Jian Yang
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Yuqi Guo
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Cuijie Lu
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Ruohan Zhang
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Yaoyu Wang
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Liang Luo
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Yanli Zhang
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Catherine H Chu
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Katherine J Wang
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Sabrine Obbad
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Wenbo Yan
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Xin Li
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA. .,Department of Urology, New York University Langone Medical Center, New York, NY, 10016, USA. .,Perlmutter Cancer Institute, New York University Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
133
|
Zhang Y, Wang H, Ren C, Yu H, Fang W, Zhang N, Gao S, Hou Q. Correlation Between C-MYC, BCL-2, and BCL-6 Protein Expression and Gene Translocation as Biomarkers in Diagnosis and Prognosis of Diffuse Large B-cell Lymphoma. Front Pharmacol 2019; 9:1497. [PMID: 30666200 PMCID: PMC6330311 DOI: 10.3389/fphar.2018.01497] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/07/2018] [Indexed: 12/22/2022] Open
Abstract
This study investigates the protein expression of C-MYC, BCL-2, and BCL-6 in diffuse large B-cell lymphoma (DLBCL) and their relationship with genetic abnormalities. A retrospective study of 42 cases on paraffin-embedded tissue specimens diagnosed with DLBCL was performed using immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH). The expression of C-MYC, BCL-2, BCL-6 protein, and gene abnormalities in these tissue samples was analyzed. The relationship in genetic abnormalities and Ki-67, Hans classification, gender, and age was also evaluated. It was found that the positive rate of C-MYC expression was 47.6% (20/42), the rate of C-MYC gene abnormality was 26.2% (11/42), in which gene translocation accounted for 23.8% (10/42) and gene amplification 2.4% (1/42); C-MYC protein expression was positively correlated with C-MYC gene translocation (χ2 = 11.813; P = 0.001); C-MYC gene translocation was mainly found in germinal center B cell type (χ2 = 4.029; P = 0.045). The positive rate of BCL-2 protein expression was 85.71% (36/42), the positive rate of translocation was 42.86% (18/42) and the amplification rate was 26.19% (11/42); the overexpression of BCL-2 protein was correlated with the BCL-2 translocation (χ2 = 3.407; P = 0.029). The positive rate of BCL-6 protein expression was 45.24% (19/42), the positive rate of BCL-6 translocation was 14.29% (6/42) and the positive rate of BCL-6 amplification was 7.14% (3/42); the overexpression of BCL-6 protein was significantly correlated with BCL-6 translocation (χ2 = 6.091; P = 0.014). The Ki-67 index was significantly higher in C-MYC translocation cases than in non-C-MYC translocation cases (χ2 = 4.492; P = 0.034). Taken together, our results suggest that the protein expression of C-MYC, BCL-2, and BCL-6 are positively correlated with their gene translocation. Overexpression of C-MYC, BCL-2, BCL-6 protein suggests the possibility of translocation. Therefore, immunohistochemical detection of C-MYC, BCL-2, and BCL-6 are useful in diagnosis and prognosis of DLBCL.
Collapse
Affiliation(s)
- YunXiang Zhang
- Department of Pathology, Weifang People's Hospital, Weifang, China
| | - Hui Wang
- Department of Pathology, Weifang People's Hospital, Weifang, China
| | - Cuiai Ren
- Department of Pathology, Weifang People's Hospital, Weifang, China
| | - Hai Yu
- Department of Pathology, Werfang Traditional Chinese Hospital, Weifang, China
| | - Wenjia Fang
- Department of Clinical Medicine, Nanchang University Medical College, Nanchang, China
| | - Na Zhang
- Department of Pathology, Weifang People's Hospital, Weifang, China
| | - Sumei Gao
- Department of Pathology, Weifang People's Hospital, Weifang, China
| | - Qian Hou
- Department of Pathology, Weifang People's Hospital, Weifang, China
| |
Collapse
|
134
|
Abstract
Comprehensive knowledge of the normal prostate epithelial lineage hierarchy is a prerequisite to investigate the identity of the cells of origin for prostate cancer. The basal and luminal cells constitute most of the prostate epithelium and have been the major focuses of the study on the cells of origin for prostate cancer. Much progress has been made during the past few decades, mainly using mouse models, to understand the inter-lineage relationship and intra-lineage heterogeneity in adults as well as the lineage plasticity during conditions of stress. These studies have concluded that the adult mouse prostate basal and luminal cells are largely independently sustained under physiological conditions, but both types of cells possess the capacity for bipotent differentiation under stress or artificial experimental conditions. However, the existence or the identity of the putative progenitors within each lineage warrants further investigation. Whether the human prostate lineage hierarchy is completely the same as that of the mouse remains uncertain. Experiments from independent groups have demonstrated that both types of cells in mice and humans can serve as targets for transformation. But controversies remain whether the disease from distinct cells of origin display different clinical behaviors. Further investigation of the intra-lineage heterogeneity will provide new insights into this issue. Understanding the identity of the cells of origin for prostate cancer will help identify novel prognostic markers for early detection of aggressive prostate cancers, provide insights into the therapeutic vulnerability of these tumors, and inspire novel therapeutic strategies.
Collapse
|
135
|
Sfanos KS, Yegnasubramanian S, Nelson WG, Lotan TL, Kulac I, Hicks JL, Zheng Q, Bieberich CJ, Haffner MC, De Marzo AM. If this is true, what does it imply? How end-user antibody validation facilitates insights into biology and disease. Asian J Urol 2019; 6:10-25. [PMID: 30775245 PMCID: PMC6363603 DOI: 10.1016/j.ajur.2018.11.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 12/30/2022] Open
Abstract
Antibodies are employed ubiquitously in biomedical sciences, including for diagnostics and therapeutics. One of the most important uses is for immunohistochemical (IHC) staining, a process that has been improving and evolving over decades. IHC is useful when properly employed, yet misuse of the method is widespread and contributes to the "reproducibility crisis" in science. We report some of the common problems encountered with IHC assays, and direct readers to a wealth of literature documenting and providing some solutions to this problem. We also describe a series of vignettes that include our approach to analytical validation of antibodies and IHC assays that have facilitated a number of biological insights into prostate cancer and the refutation of a controversial association of a viral etiology in gliomas. We postulate that a great deal of the problem with lack of accuracy in IHC assays stems from the lack of awareness by researchers for the critical necessity for end-users to validate IHC antibodies and assays in their laboratories, regardless of manufacturer claims or past publications. We suggest that one reason for the pervasive lack of end-user validation for research antibodies is that researchers fail to realize that there are two general classes of antibodies employed in IHC. First, there are antibodies that are "clinical grade" reagents used by pathologists to help render diagnoses that influence patient treatment. Such diagnostic antibodies, which tend to be highly validated prior to clinical implementation, are in the vast minority (e.g. < 500). The other main class of antibodies are "research grade" antibodies (now numbering >3 800 000), which are often not extensively validated prior to commercialization. Given increased awareness of the problem, both the United States, National Institutes of Health and some journals are requiring investigators to provide evidence of specificity of their antibody-based assays.
Collapse
Affiliation(s)
- Karen S. Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - William G. Nelson
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ibrahim Kulac
- Department of Pathology, Koc Universitesi Tip Fakultesi, Istanbul, Turkey
| | - Jessica L. Hicks
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qizhi Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles J. Bieberich
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Michael C. Haffner
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
136
|
Sollazzo M, Genchi C, Paglia S, Di Giacomo S, Pession A, de Biase D, Grifoni D. High MYC Levels Favour Multifocal Carcinogenesis. Front Genet 2018; 9:612. [PMID: 30619451 PMCID: PMC6297171 DOI: 10.3389/fgene.2018.00612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/20/2018] [Indexed: 02/05/2023] Open
Abstract
The term "field cancerisation" describes the formation of tissue sub-areas highly susceptible to multifocal tumourigenesis. In the earlier stages of cancer, cells may indeed display a series of molecular alterations that allow them to proliferate faster, eventually occupying discrete tissue regions with irrelevant morphological anomalies. This behaviour recalls cell competition, a process based on a reciprocal fitness comparison: when cells with a growth advantage arise in a tissue, they are able to commit wild-type neighbours to death and to proliferate at their expense. It is known that cells expressing high MYC levels behave as super-competitors, able to kill and replace less performant adjacent cells; given MYC upregulation in most human cancers, MYC-mediated cell competition is likely to pioneer field cancerisation. Here we show that MYC overexpression in a sub-territory of the larval wing epithelium of Drosophila is sufficient to trigger a number of cellular responses specific to mammalian pre-malignant tissues. Moreover, following induction of different second mutations, high MYC-expressing epithelia were found to be susceptible to multifocal growth, a hallmark of mammalian pre-cancerous fields. In summary, our study identified an early molecular alteration implicated in field cancerisation and established a genetically amenable model which may help study the molecular basis of early carcinogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniela Grifoni
- Cancer Evolution Laboratory, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
137
|
Abstract
Prostate cancer development involves corruption of the normal prostate transcriptional network, following deregulated expression or mutation of key transcription factors. Here, we provide an overview of the transcription factors that are important in normal prostate homeostasis (NKX3-1, p63, androgen receptor [AR]), primary prostate cancer (ETS family members, c-MYC), castration-resistant prostate cancer (AR, FOXA1), and AR-independent castration-resistant neuroendocrine prostate cancer (RB1, p53, N-MYC). We use functional (in vitro and in vivo) as well as clinical data to discuss evidence that unveils their roles in the initiation and progression of prostate cancer, with an emphasis on results of chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq).
Collapse
Affiliation(s)
- David P Labbé
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215
| |
Collapse
|
138
|
Boldrini L, Bartoletti R, Giordano M, Manassero F, Selli C, Panichi M, Galli L, Farci F, Faviana P. C-MYC, HIF-1α, ERG, TKT, and GSTP1: an Axis in Prostate Cancer? Pathol Oncol Res 2018; 25:1423-1429. [PMID: 30357756 DOI: 10.1007/s12253-018-0479-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022]
Abstract
To analyze putative biomarkers for prostate cancer (PCA) characterization, the second leading cause of cancer-associated mortality in men. Quantification of the expression level of c-myc and HIF-1α was performed in 72 prostate cancer specimens. A cohort of 497 prostate cancer patients from The Cancer Genome Atlas (TCGA) database was further analyzed, in order to test our hypothesis. We found that high c-myc level was significantly associated with HIF-1α elevated expression (p = 0.008) in our 72 samples. Statistical analysis of 497 TCGA prostate cancer specimens confirmed the strong association (p = 0.0005) of c-myc and HIF-1α expression levels, as we found in our series. Moreover, we found high c-myc levels significantly associated with low Glutatione S-transferase P1 (GSTP1) expression (p = 0.01), with high Transketolase (TKT) expression (p < 0.0001). High TKT levels were found in TCGA samples with low GSTP1 mRNA (p < 0.0001), as shown for c-myc, and with ERG increased expression (p = 0.02). Finally, samples with low GSTP1 expression displayed higher ERG mRNA levels than samples with high GSTP1 score (p < 0.0001), as above shown for c-myc. Our study emphasizes the notion of a potential value of HIF-1α and c-myc as putative biomarkers in prostate cancer; moreover TCGA data analysis showed a putative crosstalk between c-myc, HIF-1α, ERG, TKT, and GSTP1, suggesting a potential use of this axis in prostate cancer.
Collapse
Affiliation(s)
- L Boldrini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Roma 57, 56126, Pisa, Italy.
| | - R Bartoletti
- Department of Translational Research and New Technologies, University of Pisa, Pisa, Italy
| | - M Giordano
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Roma 57, 56126, Pisa, Italy
| | - F Manassero
- Division of Urology, Pisa University, Pisa, Italy
| | - C Selli
- Department of Translational Research and New Technologies, University of Pisa, Pisa, Italy
| | - M Panichi
- Department of Radiotherapy, Pisa University, Pisa, Italy
| | - L Galli
- Division of Medical Oncology, Pisa University, Pisa, Italy
| | - F Farci
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Roma 57, 56126, Pisa, Italy
| | - P Faviana
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Roma 57, 56126, Pisa, Italy
| |
Collapse
|
139
|
Guedes LB, Morais CL, Fedor H, Hicks J, Gurel B, Melamed J, Lee P, Gopalan A, Knudsen BS, True LD, Scher HI, Fine SW, Trock BJ, De Marzo AM, Lotan TL. Effect of Preanalytic Variables on an Automated PTEN Immunohistochemistry Assay for Prostate Cancer. Arch Pathol Lab Med 2018; 143:338-348. [DOI: 10.5858/arpa.2018-0068-oa] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
Phosphatase and tensin homolog (PTEN) is a promising prognostic and potentially predictive biomarker in prostate cancer.
Objective.—
To assess the effects of preanalytic variables on an analytically validated and fully automated PTEN immunohistochemistry assay.
Design.—
PTEN immunohistochemistry was performed on Ventana immunostaining systems. In benign prostate tissues, immunostaining intensity across variable conditions was assessed by digital image analysis. In prostate tumor tissues, immunostaining was scored visually.
Results.—
Delay of fixation for 4 hours or longer at room temperature or 48 hours or longer at 4°C and duration of formalin fixation did not significantly alter immunostaining intensity. Intensity of staining was highest in 10% formalin compared with other fixatives. Tumor tissues with PTEN loss processed using protocols from 11 academic institutions were all evaluable and scored identically. PTEN immunostaining of needle biopsies where tissue blocks had been stored for less than 10 years was more frequently scored as nonevaluable compared with blocks that had been stored for 10 years or longer. This effect was less evident for radical prostatectomy specimens, where low rates of nonevaluable staining were seen for 23 years or more of storage. Storage of unstained slides for 5 years at room temperature prior to immunostaining resulted in equivalent scoring compared with freshly cut slides. Machine-to-machine variability assessed across 3 Ventana platforms and 2 institutions was negligible in 12 tumors, and platform-to-platform variability was also minor comparing Ventana and Leica instruments across 77 tumors (κ = 0.926).
Conclusions.—
Automated PTEN immunostaining is robust to most preanalytic variables in the prostate and may be performed on prostate tumor tissues subjected to a wide range of preanalytic conditions. These data may help guide assay development if PTEN becomes a key predictive biomarker.
Collapse
Affiliation(s)
- Liana B. Guedes
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Carlos L. Morais
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Helen Fedor
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Jessica Hicks
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Bora Gurel
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Jonathan Melamed
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Peng Lee
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Anuradha Gopalan
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Beatrice S. Knudsen
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Lawrence D. True
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Howard I. Scher
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Samson W. Fine
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Bruce J. Trock
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Angelo M. De Marzo
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Tamara L. Lotan
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| |
Collapse
|
140
|
Zadra G, Loda M. Metabolic Vulnerabilities of Prostate Cancer: Diagnostic and Therapeutic Opportunities. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a030569. [PMID: 29229664 DOI: 10.1101/cshperspect.a030569] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer cells hijack metabolic pathways to support bioenergetics and biosynthetic requirements for their uncontrolled growth. Thus, cancer can be considered as a metabolic disease. In this review, we discuss the main metabolic features of prostate cancer with a particular focus on the link between oncogene-directed cancer metabolic regulation, metabolism rewiring, and epigenetic regulation. The potential of using metabolic profiling as a means to predict disease behavior and to identify novel therapeutic targets and new diagnostic markers will be addressed as well as the current challenges in metabolomics analyses. Finally, diagnostic and prognostic metabolic imaging approaches, including positron emission tomography, mass spectrometry, nuclear magnetic resonance, and their translational applications, will be discussed. Here, we emphasize how targeting metabolic vulnerabilities in prostate cancer may pave the way for novel personalized diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Giorgia Zadra
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215.,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215.,The Broad Institute, Cambridge, Massachusetts 02142
| |
Collapse
|
141
|
Gevaert T, Montironi R, Lopez-Beltran A, Van Leenders G, Allory Y, De Ridder D, Claessens F, Kockx M, Akand M, Joniau S, Netto G, Libbrecht L. Genito-urinary genomics and emerging biomarkers for immunomodulatory cancer treatment. Semin Cancer Biol 2018; 52:216-227. [DOI: 10.1016/j.semcancer.2017.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 01/01/2023]
|
142
|
Carneiro A, Barbosa ÁRG, Takemura LS, Kayano PP, Moran NKS, Chen CK, Wroclawski ML, Lemos GC, da Cunha IW, Obara MT, Tobias-Machado M, Sowalsky AG, Bianco B. The Role of Immunohistochemical Analysis as a Tool for the Diagnosis, Prognostic Evaluation and Treatment of Prostate Cancer: A Systematic Review of the Literature. Front Oncol 2018; 8:377. [PMID: 30280090 PMCID: PMC6153326 DOI: 10.3389/fonc.2018.00377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/23/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Prostate cancer (PCa) is a heterogeneous disease that lends itself toward numerous therapeutic options depending on its risk stratification. One of the greatest challenges in PCa urologic practice is to select patients who should be referred for biopsy and, for those patients who are diagnosed with cancer, to differentiate between patients with indolent disease from those with an unfavorable prognosis and, to determine ideal patient management and avoid unnecessary interventions. Accordingly, there is a growing body of literature reporting immunohistochemical studies with the objective of determining a prostate cancer prognosis. Among the most frequent biomarkers studied are Ki-67, p53, PTEN, MYC, and ERG. Based on these findings, we systematically reviewed articles that assessed the role of these main prognostic markers in prostate cancer. Methods: Consistent with PRISMA guidelines, we performed a systematic literature search throughout the Web of Science and PubMed Medline databases. We considered all types of studies evaluating the role of Ki-67, p53, PTEN, MYC, and ERG immunohistochemical analysis in prostate cancer until July 2017. Results: We identified 361 articles, 44 of which were summarized in this review. Diagnostically, no single immunohistochemical marker was able to define a tumor as benign or malignant. Prognostically, Ki-67, p53, and MYC were related to the tumor grade given by Gleason score and to the tumor stage (higher levels related to higher tumor grade). Furthermore, Ki-67 was also related to higher PSA levels, shorter disease-free intervals and shorter tumor-specific survival; the latter was also related to p53. The loss of PTEN protein expression showed a higher association with biochemical recurrence and with a worse prognosis, beyond that predicted by the Gleason score and tumor stage. ERG staining also showed a strong association with biochemical recurrence. Conclusion: There are several studies relating immunohistochemical markers with clinical-laboratorial outcomes in prostate cancer, the most frequent being Ki-67, p53, ERG, PTEN, and MYC. However, none of these markers have been validated by literary consensus to be routinely applied in medical practice.
Collapse
Affiliation(s)
- Arie Carneiro
- Department of Urology, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Department of Pathology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | | | | | - Carolina Ko Chen
- Department of Urology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Marcelo Langer Wroclawski
- Department of Urology, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Department of Pathology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | - Marcos Takeo Obara
- Department of Pathology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Bianca Bianco
- Human Reproduction and Genetics Center, Faculdade de Medicina do ABC, Santo André, Brazil
| |
Collapse
|
143
|
Angeles AK, Bauer S, Ratz L, Klauck SM, Sültmann H. Genome-Based Classification and Therapy of Prostate Cancer. Diagnostics (Basel) 2018; 8:E62. [PMID: 30200539 PMCID: PMC6164491 DOI: 10.3390/diagnostics8030062] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/19/2022] Open
Abstract
In the past decade, multi-national and multi-center efforts were launched to sequence prostate cancer genomes, transcriptomes, and epigenomes with the aim of discovering the molecular underpinnings of tumorigenesis, cancer progression, and therapy resistance. Multiple biological markers and pathways have been discovered to be tumor drivers, and a molecular classification of prostate cancer is emerging. Here, we highlight crucial findings of these genome-sequencing projects in localized and advanced disease. We recapitulate the utility and limitations of current clinical practices to diagnosis, prognosis, and therapy, and we provide examples of insights generated by the molecular profiling of tumors. Novel treatment concepts based on these molecular alterations are currently being addressed in clinical trials and will lead to an enhanced implementation of precision medicine strategies.
Collapse
Affiliation(s)
- Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| | - Simone Bauer
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| | - Leonie Ratz
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg D-69120, Germany.
| |
Collapse
|
144
|
Ciccarelli C, Di Rocco A, Gravina GL, Mauro A, Festuccia C, Del Fattore A, Berardinelli P, De Felice F, Musio D, Bouché M, Tombolini V, Zani BM, Marampon F. Disruption of MEK/ERK/c-Myc signaling radiosensitizes prostate cancer cells in vitro and in vivo. J Cancer Res Clin Oncol 2018; 144:1685-1699. [PMID: 29959569 DOI: 10.1007/s00432-018-2696-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/26/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Prostate cancer (PCa) cell radioresistance causes the failure of radiation therapy (RT) in localized or locally advanced disease. The aberrant accumulation of c-Myc oncoprotein, known to promote PCa onset and progression, may be due to the control of gene transcription and/or MEK/ERK-regulated protein stabilization. Here, we investigated the role of MEK/ERK signaling in PCa. METHODS LnCAP, 22Rv1, DU145, and PC3 PCa cell lines were used in in vitro and in vivo experiments. U0126, trametinib MEK/ERK inhibitors, and c-Myc shRNAs were used. Radiation was delivered using an x-6 MV photon linear accelerator. U0126 in vivo activity alone or in combination with irradiation was determined in murine xenografts. RESULTS Inhibition of MEK/ERK signaling down-regulated c-Myc protein in PCa cell lines to varying extents by affecting expression of RNA and protein, which in turn determined radiosensitization in in vitro and in vivo xenograft models of PCa cells. The crucial role played by c-Myc in the MEK/ERK pathways was demonstrated in 22Rv1 cells by the silencing of c-Myc by means of short hairpin mRNA, which yielded effects resembling the targeting of MEK/ERK signaling. The clinically approved compound trametinib used in vitro yielded the same effects as U0126 on growth and C-Myc expression. Notably, U0126 and trametinib induced a drastic down-regulation of BMX, which is known to prevent apoptosis in cancer cells. CONCLUSIONS The results of our study suggest that signal transduction-based therapy can, by disrupting the MEK/ERK/c-Myc axis, reduce human PCa radioresistance caused by increased c-Myc expression in vivo and in vitro and restores apoptosis signals.
Collapse
Affiliation(s)
- Carmela Ciccarelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Agnese Di Rocco
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Annunziata Mauro
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy
| | - Andrea Del Fattore
- Multi-Factorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, IRCCS, Viale di San Paolo 15, 00146, Rome, Italy
| | - Paolo Berardinelli
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Francesca De Felice
- Division of Radiotherapy, Department of Radiology, Radiation Oncology and Human Pathology, "Sapienza" University of Rome, Rome, Italy
| | - Daniela Musio
- Division of Radiotherapy, Department of Radiology, Radiation Oncology and Human Pathology, "Sapienza" University of Rome, Rome, Italy
| | - Marina Bouché
- Unit of Histology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Tombolini
- Unit of Histology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Bianca Maria Zani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
- Unit of Histology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
145
|
Chellini L, Monteleone V, Lombari M, Caldarola S, Loreni F. The oncoprotein Myc controls the phosphorylation of S6 kinase and AKT through protein phosphatase 2A. J Cell Biochem 2018; 119:9878-9887. [PMID: 30132971 DOI: 10.1002/jcb.27309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 06/27/2018] [Indexed: 12/29/2022]
Abstract
This study focuses on the effects of Myc oncoprotein on the translational apparatus of the cell. Translation is an energy consuming process that involves a large number of accessory factors. The production of components of the protein synthesis machinery can be regulated at the transcriptional level by specific factors. It has been shown that the product of the oncogene Myc, a transcription factor frequently activated in cancer, can control translational activity through an increase in the transcription of the eIF4F complex components (eIF4E, eIF4AI, and eIF4GI). However, additional effects at the posttranslational level have also been described. For instance, it has been shown that Myc upregulation can induce mammalian target of rapamycin (mTOR)-dependent 4E-binding protein 1 (4E-BP1) hyperphosphorylation. We induced overexpression or inhibition of Myc through transfection of complementary DNA constructs or specific small interfering RNA in PC3 (prostate carcinoma) and HeLa (cervical carcinoma) cells. We have observed that overexpression of Myc causes an increase in 4E-BP1 phosphorylation and activation of protein synthesis. Unexpectedly, we detected a parallel decrease in the phosphorylation level of S6 kinase (in PC3 and HeLa) and AKT (in HeLa). We report evidence that these changes are mediated by an increase in protein phosphatase 2A activity.
Collapse
Affiliation(s)
- Lidia Chellini
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.,Unit of Preclinical Models and New Therapeutic Agents, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | | | - Malinska Lombari
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Sara Caldarola
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Fabrizio Loreni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
146
|
Lin TP, Li J, Li Q, Li X, Liu C, Zeng N, Huang JM, Chu GCY, Lin CH, Zhau HE, Chung LWK, Wu BJ, Shih JC. R1 Regulates Prostate Tumor Growth and Progression By Transcriptional Suppression of the E3 Ligase HUWE1 to Stabilize c-Myc. Mol Cancer Res 2018; 16:1940-1951. [PMID: 30042175 DOI: 10.1158/1541-7786.mcr-16-0346] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/22/2018] [Accepted: 07/05/2018] [Indexed: 11/16/2022]
Abstract
Prostate cancer is a prevalent public health problem, especially because noncutaneous advanced malignant forms significantly affect the lifespan and quality of life of men worldwide. New therapeutic targets and approaches are urgently needed. The current study reports elevated expression of R1 (CDCA7L/RAM2/JPO2), a c-Myc-interacting protein and transcription factor, in human prostate cancer tissue specimens. In a clinical cohort, high R1 expression is associated with disease recurrence and decreased patient survival. Overexpression and knockdown of R1 in human prostate cancer cells indicate that R1 induces cell proliferation and colony formation. Moreover, silencing R1 dramatically reduces the growth of prostate tumor xenografts in mice. Mechanistically, R1 increases c-Myc protein stability by inhibiting ubiquitination and proteolysis through transcriptional suppression of HUWE1, a c-Myc-targeting E3 ligase, via direct interaction with a binding element in the promoter. Moreover, transcriptional repression is supported by a negative coexpression correlation between R1 and HUWE1 in a prostate cancer clinical dataset. Collectively, these findings, for the first time, characterize the contribution of R1 to prostate cancer pathogenesis. IMPLICATIONS: These findings provide evidence that R1 is a novel regulator of prostate tumor growth by stabilizing c-Myc protein, meriting further investigation of its therapeutic and prognostic potential.
Collapse
Affiliation(s)
- Tzu-Ping Lin
- Depatment of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
- USC-Taiwan Center for Translational Research, University of Southern California, Los Angeles, California
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Urology, School of Medicine and Shu-Tien Urological Research Center, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Jingjing Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Qinlong Li
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Pathology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiangyan Li
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Chunyan Liu
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ni Zeng
- Depatment of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Jen-Ming Huang
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Gina Chia-Yi Chu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Chi-Hung Lin
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Leland W K Chung
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington.
| | - Jean C Shih
- Depatment of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California.
- USC-Taiwan Center for Translational Research, University of Southern California, Los Angeles, California
- Depatment of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
147
|
Qu H, Liu H, Jin Y, Cui Z, Han G. HUWE1 upregulation has tumor suppressive effect in human prostate cancer cell lines through c-Myc. Biomed Pharmacother 2018; 106:309-315. [PMID: 29966975 DOI: 10.1016/j.biopha.2018.06.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/07/2018] [Accepted: 06/12/2018] [Indexed: 01/23/2023] Open
Abstract
PURPOSE We investigated the regulatory function of HECT, UBA and WWE domain-containing protein 1, E3 ubiquitin protein ligase (HUWE1) in human prostate cancer (CaP). METHODS HUWE1 was overexpressed (through transfection) or downregulated (through lentiviral transduction) in CaP cell lines, PC3 and DU145 cells. The functions of HUWE1 overexpression or downregulation on CaP cancer cell proliferation, migrationin vitro, and explant in vivo were examined. In addition, the regulatory effect of HUWE1 on c-Myc expression was assessed. In HUWE1-overexpressed CaP cells, c-Myc was further upregulated to assess whether c-Myc was directly involved in HUWE1-induced regulation in CaP. RESULTS HUWE1 overexpression inhibited CaP proliferation and migrationin vitro, and explant growth in vivo. On the other hand, HUWE1 downregulation had no effects on CaP in vitro. C-Myc was downregulated in HUWE1-overexpressed, but un-changed in HUWE1-downregulated, CaP cells. Further upregulating c-Myc in HUWE1-overexpressed CaP cells reversed the tumor-suppressing effects by HUWE1-overexpression on cancer proliferation and migration in vitro. CONCLUSION HUWE1 overexpression could functionally suppress CaP development bothin vitro and in vivo, possibly by inverse regulation on c-Myc.
Collapse
Affiliation(s)
- Huawei Qu
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, China; School of Medicine, Shandong University, Jinan, 250012, China
| | - Hui Liu
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, China
| | - Yang Jin
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, China
| | - Zilian Cui
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, China
| | - Gang Han
- Department of Urology, Chinese PLA 252 Hospital, Baoding, 071000, China.
| |
Collapse
|
148
|
PanCD44 Immunohistochemical Evaluation in Prostatectomies from Patients with Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2061268. [PMID: 29682524 PMCID: PMC5846379 DOI: 10.1155/2018/2061268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 12/03/2017] [Accepted: 01/15/2018] [Indexed: 01/30/2023]
Abstract
Introduction CD44 has been proposed as a prognostic marker and a stem cell marker but studies in patients with prostate cancer have yielded inconsistent results. Patients and Methods Patients submitted to radical prostatectomy between 2008 and 2013 at a university hospital were followed with biannual serum PSA tests to determine the biochemical recurrence (BR). Archived paraffin blocks with neoplastic and nonneoplastic tissue were evaluated immunohistochemically for a panCD44 and MYC. Results Sixty-nine patients completed follow-up and were included. CD44 positivity was observed in inflammatory cells (42%), nonneoplastic epithelium (39.7%), and neoplastic tissue (12.3%). In nonneoplastic tissues staining was observed in basal and luminal cells with the morphology of terminally differentiated cells. In neoplastic tissues, CD44 negativity was correlated with higher Gleason scores (Rho = −0.204; p = 0.042) and higher preoperative serum PSA levels when evaluated continuously (p = 0.029). CD44 expression was not associated with tumor stage (p = 0.668), surgical margin status (p = 0.471), or BR (p = 0.346), nor was there any association between CD44 and MYC expression in neoplastic tissue (p = 1.0). Conclusion In the bulk of cells, the minority of cancer stem cells would not be detected by immunohistochemistry using panCD44. As a prognostic marker, its expression was weakly correlated with Gleason score and preoperative PSA level, but not with surgical margin status, tumor stage, or BR.
Collapse
|
149
|
Prostate Cancer Genomics: Recent Advances and the Prevailing Underrepresentation from Racial and Ethnic Minorities. Int J Mol Sci 2018; 19:ijms19041255. [PMID: 29690565 PMCID: PMC5979433 DOI: 10.3390/ijms19041255] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/15/2018] [Accepted: 04/15/2018] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed non-cutaneous cancer and the second leading cause of male cancer deaths in the United States. Among African American (AA) men, CaP is the most prevalent malignancy, with disproportionately higher incidence and mortality rates. Even after discounting the influence of socioeconomic factors, the effect of molecular and genetic factors on racial disparity of CaP is evident. Earlier studies on the molecular basis for CaP disparity have focused on the influence of heritable mutations and single-nucleotide polymorphisms (SNPs). Most CaP susceptibility alleles identified based on genome-wide association studies (GWAS) were common, low-penetrance variants. Germline CaP-associated mutations that are highly penetrant, such as those found in HOXB13 and BRCA2, are usually rare. More recently, genomic studies enabled by Next-Gen Sequencing (NGS) technologies have focused on the identification of somatic mutations that contribute to CaP tumorigenesis. These studies confirmed the high prevalence of ERG gene fusions and PTEN deletions among Caucasian Americans and identified novel somatic alterations in SPOP and FOXA1 genes in early stages of CaP. Individuals with African ancestry and other minorities are often underrepresented in these large-scale genomic studies, which are performed primarily using tumors from men of European ancestry. The insufficient number of specimens from AA men and other minority populations, together with the heterogeneity in the molecular etiology of CaP across populations, challenge the generalizability of findings from these projects. Efforts to close this gap by sequencing larger numbers of tumor specimens from more diverse populations, although still at an early stage, have discovered distinct genomic alterations. These research findings can have a direct impact on the diagnosis of CaP, the stratification of patients for treatment, and can help to address the disparity in incidence and mortality of CaP. This review examines the progress of understanding in CaP genetics and genomics and highlight the need to increase the representation from minority populations.
Collapse
|
150
|
Calcium and Nuclear Signaling in Prostate Cancer. Int J Mol Sci 2018; 19:ijms19041237. [PMID: 29671777 PMCID: PMC5979488 DOI: 10.3390/ijms19041237] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/15/2018] [Accepted: 04/17/2018] [Indexed: 02/06/2023] Open
Abstract
Recently, there have been a number of developments in the fields of calcium and nuclear signaling that point to new avenues for a more effective diagnosis and treatment of prostate cancer. An example is the discovery of new classes of molecules involved in calcium-regulated nuclear import and nuclear calcium signaling, from the G protein-coupled receptor (GPCR) and myosin families. This review surveys the new state of the calcium and nuclear signaling fields with the aim of identifying the unifying themes that hold out promise in the context of the problems presented by prostate cancer. Genomic perturbations, kinase cascades, developmental pathways, and channels and transporters are covered, with an emphasis on nuclear transport and functions. Special attention is paid to the molecular mechanisms behind prostate cancer progression to the malignant forms and the unfavorable response to anti-androgen treatment. The survey leads to some new hypotheses that connect heretofore disparate results and may present a translational interest.
Collapse
|