1
|
Stopsack KH, Huang Y, Tyekucheva S, Gerke TA, Bango C, Elfandy H, Bowden M, Penney KL, Roberts TM, Parmigiani G, Kantoff PW, Mucci LA, Loda M. Multiplex Immunofluorescence in Formalin-Fixed Paraffin-Embedded Tumor Tissue to Identify Single-Cell-Level PI3K Pathway Activation. Clin Cancer Res 2020; 26:5903-5913. [PMID: 32913135 DOI: 10.1158/1078-0432.ccr-20-2000] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/11/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Identifying cancers with high PI3K pathway activity is critical for treatment selection and eligibility into clinical trials of PI3K inhibitors. Assessments of tumor signaling pathway activity need to consider intratumoral heterogeneity and multiple regulatory nodes. EXPERIMENTAL DESIGN We established a novel, mechanistically informed approach to assessing tumor signaling pathways by quantifying single-cell-level multiplex immunofluorescence using custom algorithms. In a proof-of-concept study, we stained archival formalin-fixed, paraffin-embedded (FFPE) tissue from patients with primary prostate cancer in two prospective cohort studies, the Health Professionals Follow-up Study and the Physicians' Health Study. PTEN, stathmin, and phospho-S6 were quantified on 14 tissue microarrays as indicators of PI3K activation to derive cell-level PI3K scores. RESULTS In 1,001 men, 988,254 tumor cells were assessed (median, 743 per tumor; interquartile range, 290-1,377). PI3K scores were higher in tumors with PTEN loss scored by a pathologist, higher Gleason grade, and a new, validated bulk PI3K transcriptional signature. Unsupervised machine-learning approaches resulted in similar clustering. Within-tumor heterogeneity in cell-level PI3K scores was high. During long-term follow-up (median, 15.3 years), rates of progression to metastases and death from prostate cancer were twice as high in the highest quartile of PI3K activation compared with the lowest quartile (hazard ratio, 2.04; 95% confidence interval, 1.13-3.68). CONCLUSIONS Our novel pathway-focused approach to quantifying single-cell-level immunofluorescence in FFPE tissue identifies prostate tumors with PI3K pathway activation that are more aggressive and may respond to pathway inhibitors.
Collapse
Affiliation(s)
- Konrad H Stopsack
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ying Huang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Svitlana Tyekucheva
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Travis A Gerke
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Clyde Bango
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Habiba Elfandy
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Michaela Bowden
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Thomas M Roberts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Giovanni Parmigiani
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. .,Department of Pathology, Weill Cornell Medical College, New York, New York.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,New York Genome Center, New York, New York
| |
Collapse
|
2
|
Khan S, Caldwell J, Wilson KM, Gonzalez-Feliciano AG, Peisch S, Pernar CH, Graff RE, Giovannucci EL, Mucci LA, Gerke TA, Markt SC. Baldness and Risk of Prostate Cancer in the Health Professionals Follow-up Study. Cancer Epidemiol Biomarkers Prev 2020; 29:1229-1236. [PMID: 32277004 DOI: 10.1158/1055-9965.epi-19-1236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/07/2019] [Accepted: 04/03/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The association between male pattern baldness and prostate cancer has been inconsistent. We prospectively investigated the association between baldness at age 45 and prostate cancer risk in the Health Professionals Follow-up Study (HPFS), focusing on clinical and molecular markers. METHODS Baldness was self-reported on the 1992 questionnaire using the modified Norwood-Hamilton scale prior to diagnosis. We estimated HRs between baldness and prostate cancer risk among 36,760 men, with follow-up through 2014. We also investigated whether baldness was associated with prostate cancer defined by tumor protein expression of androgen receptor and the presence of the TMPRSS2:ERG fusion. RESULTS During 22 years, 5,157 prostate cancer cases were identified. Fifty-six percent of the men had either frontal or vertex baldness. No significant associations were found between baldness and prostate cancer risk. Among men younger than 60 years, there was a statistically significant association between frontal and severe vertex baldness and overall prostate cancer (HR: 1.74; 95% confidence interval: 1.23-2.48). Baldness was not significantly associated with expression of molecular subtypes defined by AR and TMPRSS2:ERG IHC of prostate tumors. CONCLUSIONS This study showed no association between baldness at age 45 and prostate cancer risk, overall or for clinical or molecular markers. The association between baldness and overall prostate cancer among younger men is intriguing, but caution is warranted when interpreting this finding. IMPACT The null findings from this large cohort study, together with previous literature's inconclusive findings across baldness patterns, suggest that baldness is not a consistent biomarker for prostate cancer risk or progression.
Collapse
Affiliation(s)
- Saud Khan
- Liaquat University of Medical and Health Sciences, Sindh, Pakistan
| | | | - Kathryn M Wilson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Samuel Peisch
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Claire H Pernar
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Rebecca E Graff
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Sarah C Markt
- Department of Population and Quantitative Health Science, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
3
|
Wegner KA, Mueller BR, Unterberger CJ, Avila EJ, Ruetten H, Turco AE, Oakes SR, Girardi NM, Halberg RB, Swanson SM, Marker PC, Vezina CM. Prostate epithelial-specific expression of activated PI3K drives stromal collagen production and accumulation. J Pathol 2019; 250:231-242. [PMID: 31674011 DOI: 10.1002/path.5363] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/30/2019] [Accepted: 10/27/2019] [Indexed: 01/01/2023]
Abstract
We genetically engineered expression of an activated form of P110 alpha, the catalytic subunit of PI3K, in mouse prostate epithelium to create a mouse model of direct PI3K activation (Pbsn-cre4Prb;PI3KGOF/+ ). We hypothesized that direct activation would cause rapid neoplasia and cancer progression. Pbsn-cre4Prb;PI3KGOF/+ mice developed widespread prostate intraepithelial hyperplasia, but stromal invasion was limited and overall progression was slower than anticipated. However, the model produced profound and progressive stromal remodeling prior to explicit epithelial neoplasia. Increased stromal cellularity and inflammatory infiltrate were evident as early as 4 months of age and progressively increased through 12 months of age, the terminal endpoint of this study. Prostatic collagen density and phosphorylated SMAD2-positive prostatic stromal cells were expansive and accumulated with age, consistent with pro-fibrotic TGF-β pathway activation. Few reported mouse models accumulate prostate-specific collagen to the degree observed in Pbsn-cre4Prb;PI3KGOF/+ . Our results indicate a signaling process beginning with prostatic epithelial PI3K and TGF-β signaling that drives prostatic stromal hypertrophy and collagen accumulation. These mice afford a unique opportunity to explore molecular mechanisms of prostatic collagen accumulation that is relevant to cancer progression, metastasis, inflammation and urinary dysfunction. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kyle A Wegner
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Brett R Mueller
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Christopher J Unterberger
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Enrique J Avila
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Hannah Ruetten
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Anne E Turco
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Steven R Oakes
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicholas M Girardi
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard B Halberg
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA.,Division of Gastroenterology and Hepatology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Steven M Swanson
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Paul C Marker
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Chad M Vezina
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
4
|
Enabling precision medicine by unravelling disease pathophysiology: quantifying signal transduction pathway activity across cell and tissue types. Sci Rep 2019; 9:1603. [PMID: 30733525 PMCID: PMC6367506 DOI: 10.1038/s41598-018-38179-x] [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: 09/06/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022] Open
Abstract
Signal transduction pathways are important in physiology and pathophysiology. Targeted drugs aim at modifying pathogenic pathway activity, e.g., in cancer. Optimal treatment choice requires assays to measure pathway activity in individual patient tissue or cell samples. We developed a method enabling quantitative measurement of functional pathway activity based on Bayesian computational model inference of pathway activity from measurements of mRNA levels of target genes of the pathway-associated transcription factor. Oestrogen receptor, Wnt, and PI3K-FOXO pathway assays have been described previously. Here, we report model development for androgen receptor, Hedgehog, TGFβ, and NFκB pathway assays, biological validation on multiple cell types, and analysis of data from published clinical studies (multiple sclerosis, amyotrophic lateral sclerosis, contact dermatitis, Ewing sarcoma, lymphoma, medulloblastoma, ependymoma, skin and prostate cancer). Multiple pathway analysis of clinical prostate cancer (PCa) studies showed increased AR activity in hyperplasia and primary PCa but variable AR activity in castrate resistant (CR) PCa, loss of TGFβ activity in PCa, increased Wnt activity in TMPRSS2:ERG fusion protein-positive PCa, active PI3K pathway in advanced PCa, and active PI3K and NFκB as potential hormonal resistance pathways. Potential value for future clinical practice includes disease subtyping and prediction and targeted therapy response prediction and monitoring.
Collapse
|
5
|
Torrealba N, Rodriguez-Berriguete G, Fraile B, Olmedilla G, Martínez-Onsurbe P, Sánchez-Chapado M, Paniagua R, Royuela M. PI3K pathway and Bcl-2 family. Clinicopathological features in prostate cancer. Aging Male 2018; 21:211-222. [PMID: 29316844 DOI: 10.1080/13685538.2018.1424130] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR pathways and Bcl-2 family play a central role in prostate cancer (PC). The aim was to determine influence in the biochemical progression in PC. To evaluate the association between clinic pathological and immunohistochemical variables, Spearman's test was performed. Log-rank test and Kaplan-Meier curves were used for survival comparisons. To explore the correlation of the studied immunohistochemical parameters and the established prognostic variables with biochemical progression, univariate and multivariate Cox proportional Hazard regression analyses were performed. Spearman analysis showed correlation between stroma expression and tumor expression of PI3K with biochemical progression (p = .009, p = .004), respectively, and tumor immunohistochemical score with biochemical progression (p = .051). In the multivariate Cox regression model, only PI3K was retained as independent predictors of biochemical progression. In stroma expression, PI3K is (HR 0.172, 95% CI 0.065-0.452, p = .000); tumor expression, PI3K is (HR 0.087, 95% CI 0.026-0.293, p = .000), and tumor immunohistochemical score (HR 0.382, 95% CI 0.209-0.697 p = .002). Our results suggest a role for prostatic expression of PI3K was prognostic markers for PC. PI3K/AKT/mTOR and Bcl-2 family are becoming an important therapeutic target and predictive biomarkers of onset and progression of PC.
Collapse
Affiliation(s)
- Norelia Torrealba
- a Department of Biomedicine and Biotechnology , University of Alcalá , Alcalá de Henares , Spain
| | | | - Benito Fraile
- a Department of Biomedicine and Biotechnology , University of Alcalá , Alcalá de Henares , Spain
| | - Gabriel Olmedilla
- b Department of Pathology , University of Alcalá , Alcalá de Henares , Spain
| | | | | | - Ricardo Paniagua
- a Department of Biomedicine and Biotechnology , University of Alcalá , Alcalá de Henares , Spain
| | - Mar Royuela
- a Department of Biomedicine and Biotechnology , University of Alcalá , Alcalá de Henares , Spain
| |
Collapse
|
6
|
Zhang S, Li J, Zhou G, Mu D, Yan J, Xing J, Yao Z, Sheng H, Li D, Lv C, Sun B, Hong Q, Guo H. Aurora-A regulates autophagy through the Akt pathway in human prostate cancer. Cancer Biomark 2018; 19:27-34. [PMID: 28269749 DOI: 10.3233/cbm-160238] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Aurora A kinase is frequently overexpressed in a variety of tumor types, including the prostate. However, the function of Aurora A in autophagy in prostate cancer has not been investigated. Here, we aimed to study the functioning mechanism and autophagy associated signaling pathways of Aurora A in prostate cancer. METHODS To investigate the biological function of Aurora A, down-regulation of Aurora A was performed followed by functional testing assays. Immunohistochemistry was used to detect the expression of Aurora A in human prostate cancer specimens. CCK8, Transwell, flow cytometric analysis and measurement of tumor formation in nude mice were performed to test the effects of Aurora A down-regulation in vivo and in vitro. Signaling pathway analysis was performed by using Western blot. Autophagy activity was measured by monitoring the expression levels of LC3-II. RESULTS Aurora A overexpression was significantly higher in human prostate cancer specimens than in BPH. Furthermore, Aurora A knockdown inhibited the proliferation of prostate cancer cells by suppressing the Akt pathway, indicating that Akt is a novel Aurora A substrate in prostate cancer. Additionally, Aurora A down-regulation prompts autophagy in prostate cancer cells. Most importantly, Aurora A ablation almost fully abrogates tumorigenesis in nude mice, suggesting that Aurora A is a key oncogenic effector in prostate cancer. CONCLUSIONS Taken together, our data suggest that Aurora-A plays an important role in the suppression of autophagy by inhibiting the phosphorylation of Akt, which in turn prevents autophagy-induced apoptosis in prostate cancer.
Collapse
|
7
|
Ramroop JR, Stein MN, Drake JM. Impact of Phosphoproteomics in the Era of Precision Medicine for Prostate Cancer. Front Oncol 2018; 8:28. [PMID: 29503809 PMCID: PMC5820335 DOI: 10.3389/fonc.2018.00028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/29/2018] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer is the most common malignancy in men in the United States. While androgen deprivation therapy results in tumor responses initially, there is relapse and progression to metastatic castration-resistant prostate cancer. Currently, all prostate cancer patients receive essentially the same treatment, and there is a need for clinically applicable technologies to provide predictive biomarkers toward personalized therapies. Genomic analyses of tumors are used for clinical applications, but with a paucity of obvious driver mutations in metastatic castration-resistant prostate cancer, other applications, such as phosphoproteomics, may complement this approach. Immunohistochemistry and reverse phase protein arrays are limited by the availability of reliable antibodies and evaluates a preselected number of targets. Mass spectrometry-based phosphoproteomics has been used to profile tumors consisting of thousands of phosphopeptides from individual patients after surgical resection or at autopsy. However, this approach is time consuming, and while a large number of candidate phosphopeptides are obtained for evaluation, limitations are reduced reproducibility, sensitivity, and precision. Targeted mass spectrometry can help eliminate these limitations and is more cost effective and less time consuming making it a practical platform for future clinical testing. In this review, we discuss the use of phosphoproteomics in prostate cancer and other clinical cancer tissues for target identification, hypothesis testing, and possible patient stratification. We highlight the majority of studies that have used phosphoproteomics in prostate cancer tissues and cell lines and propose ways forward to apply this approach in basic and clinical research. Overall, the implementation of phosphoproteomics via targeted mass spectrometry has tremendous potential to aid in the development of more rational, personalized therapies that will result in increased survival and quality of life enhancement in patients suffering from metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Johnny R. Ramroop
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Mark N. Stein
- Developmental Therapeutics/Phase I Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- Department of Medicine, Division of Medical Oncology and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Justin M. Drake
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- Department of Medicine, Division of Medical Oncology and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
8
|
Chakravarthi BVSK, Chandrashekar DS, Agarwal S, Balasubramanya SAH, Pathi SS, Goswami MT, Jing X, Wang R, Mehra R, Asangani IA, Chinnaiyan AM, Manne U, Sonpavde G, Netto GJ, Gordetsky J, Varambally S. miR-34a Regulates Expression of the Stathmin-1 Oncoprotein and Prostate Cancer Progression. Mol Cancer Res 2017; 16:1125-1137. [PMID: 29025958 DOI: 10.1158/1541-7786.mcr-17-0230] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/24/2017] [Accepted: 10/09/2017] [Indexed: 12/27/2022]
Abstract
In aggressive prostate cancers, the oncoprotein STMN1 (also known as stathmin 1 and oncoprotein 18) is often overexpressed. STMN1 is involved in various cellular processes, including cell proliferation, motility, and tumor metastasis. Here, it was found that the expression of STMN1 RNA and protein is elevated in metastatic prostate cancers. Knockdown of STMN1 resulted in reduced proliferation and invasion of cells and tumor growth and metastasis in vivo Furthermore, miR-34a downregulated STMN1 by directly binding to its 3'-UTR. Overexpression of miR-34a in prostate cancer cells reduced proliferation and colony formation, suggesting that it is a tumor suppressor. The transcriptional corepressor C-terminal binding protein 1 (CtBP1) negatively regulated expression of miR-34a. Furthermore, gene expression profiling of STMN1-modulated prostate cancer cells revealed molecular alterations, including elevated expression of growth differentiation factor 15 (GDF15), which is involved in cancer progression and potentially in STMN1-mediated oncogenesis. Thus, in prostate cancer, CtBP1-regulated miR-34a modulates STMN1 expression and is involved in cancer progression through the CtBP1\miR-34a\STMN1\GDF15 axis.Implications: The CtBP1\miR-34a\STMN1\GDF15 axis is a potential therapeutic target for treatment of aggressive prostate cancer. Mol Cancer Res; 16(7); 1125-37. ©2017 AACR.
Collapse
Affiliation(s)
- Balabhadrapatruni V S K Chakravarthi
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Sumit Agarwal
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Satya S Pathi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Moloy T Goswami
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Xiaojun Jing
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan.,Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Rohit Mehra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan.,Department of Pathology, University of Michigan, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Irfan A Asangani
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan.,Department of Pathology, University of Michigan, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan.,Department of Urology, University of Michigan, Ann Arbor, Michigan.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Guru Sonpavde
- Department of Medical Oncology, GU section, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer Gordetsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama. .,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
9
|
Lu D, Carlsson J, Penney KL, Davidsson S, Andersson SO, Mucci LA, Valdimarsdóttir U, Andrén O, Fang F, Fall K. Expression and Genetic Variation in Neuroendocrine Signaling Pathways in Lethal and Nonlethal Prostate Cancer among Men Diagnosed with Localized Disease. Cancer Epidemiol Biomarkers Prev 2017; 26:1781-1787. [PMID: 28939587 DOI: 10.1158/1055-9965.epi-17-0453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/14/2017] [Accepted: 09/12/2017] [Indexed: 11/16/2022] Open
Abstract
Background: Recent data suggest that neuroendocrine signaling pathways may play a role in the progression of prostate cancer, particularly for early-stage disease. We aimed to explore whether expression of selected genes in the adrenergic, serotoninergic, glucocorticoid, and dopaminergic pathways differs in prostate tumor tissue from men with lethal disease compared with men with nonlethal disease.Methods: On the basis of the Swedish Watchful Waiting Cohort, we included 511 men diagnosed with incidental prostate cancer through transurethral resection of the prostate during 1977-1998 with follow-up up to 30 years. For those with tumor tissue (N = 262), we measured mRNA expression of 223 selected genes included in neuroendocrine pathways. Using DNA from normal prostate tissue (N = 396), we genotyped 36 SNPs from 14 receptor genes. Lethal prostate cancer was the primary outcome in analyses with pathway gene expression and genetic variants.Results: Differential expression of genes in the serotoninergic pathway was associated with risk of lethal prostate cancer (P = 0.007); similar but weaker associations were noted for the adrenergic (P = 0.014) and glucocorticoid (P = 0.020) pathways. Variants of the HTR2A (rs2296972; P = 0.002) and NR3CI (rs33388; P = 0.035) genes (within the serotoninergic and glucocorticoid pathways) were associated with lethal cancer in overdominant models. These genetic variants were correlated with expression of several genes in corresponding pathways (P < 0.05).Conclusions: Our findings lend support to hypothesis that the neuroendocrine pathways, particularly serotoninergic pathway, are associated with lethal outcome in the natural course of localized prostate cancer.Impact: This study provides evidence of the role of neuroendocrine pathways in prostate cancer progression that may have clinical utility. Cancer Epidemiol Biomarkers Prev; 26(12); 1781-7. ©2017 AACR.
Collapse
Affiliation(s)
- Donghao Lu
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Jessica Carlsson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sabina Davidsson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Swen-Olof Andersson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Unnur Valdimarsdóttir
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Faculty of Medicine, Center of Public Health Sciences, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| | - Ove Andrén
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Fang Fang
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Katja Fall
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
10
|
Hu J, Wang G, Sun T. Dissecting the roles of the androgen receptor in prostate cancer from molecular perspectives. Tumour Biol 2017; 39:1010428317692259. [PMID: 28475016 DOI: 10.1177/1010428317692259] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Androgen receptor plays a pivotal role in prostate cancer progression, and androgen deprivation therapy to intercept androgen receptor signal pathway is an indispensable treatment for most advanced prostate cancer patients to delay cancer progression. However, the emerging of castration-resistant prostate cancer reminds us the alteration of androgen receptor, which includes androgen receptor mutation, the formation of androgen receptor variants, and androgen receptor distribution in cancer cells. In this review, we introduce the process of androgen receptor and also its variants' formation, translocation, and function alteration by protein modification or interaction with other pathways. We dissect the roles of androgen receptor in prostate cancer from molecular perspective to provide clues for battling prostate cancer, especially castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Jieping Hu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Sun
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Chan ML, Yu CC, Hsu JL, Leu WJ, Chan SH, Hsu LC, Liu SP, Ivantcova PM, Dogan Ö, Bräse S, Kudryavtsev KV, Guh JH. Enantiomerically pure β-dipeptide derivative induces anticancer activity against human hormone-refractory prostate cancer through both PI3K/Akt-dependent and -independent pathways. Oncotarget 2017; 8:96668-96683. [PMID: 29228561 PMCID: PMC5722513 DOI: 10.18632/oncotarget.18040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/08/2017] [Indexed: 11/25/2022] Open
Abstract
The use of peptides that target cancer cells and induce anticancer activities through various mechanisms is developing as a potential anticancer strategy. KUD983, an enantiomerically pure β-dipeptide derivative, displays potent activity against hormone-refractory prostate cancer (HRPC) PC-3 and DU145 cells with submicromolar IC50. KUD983 induced G1 arrest of the cell cycle and subsequent apoptosis associated with down-regulation of several related proteins including cyclin D1, cyclin E and Cdk4, and the de-phosphorylation of RB. The levels of nuclear and total c-Myc protein, which could increase the expression of both cyclin D1 and cyclin E, were profoundly inhibited by KUD983. Furthermore, it inhibited PI3K/Akt and mTOR/p70S6K/4E-BP1 pathways, the key signaling in multiple cellular functions. The transient transfection of constitutively active myristylated Akt (myr-Akt) cDNA significantly rescued KUD983-induced caspase activation but did not blunt the inhibition of mTOR/p70S6K/4E-BP1 signaling cascade suggesting the presence of both Akt-dependent and -independent pathways. Moreover, KUD983-induced effect was enhanced with the down-regulation of anti-apoptotic Bcl-2 members (e.g., Bcl-2, and Mcl-1) and IAP family members (e.g., survivin). Notably, KUD983 induced autophagic cell death using confocal microscopic examination, tracking the level of conversion of LC3-I to LC3-II and flow cytometric detection of acidic vesicular organelles-positive cells. In conclusion, the data suggest that KUD983 is an anticancer β-dipeptide against HRPCs through the inhibition of cell proliferation and induction of apoptotic and autophagic cell death. The suppression of signaling pathways regulated by c-Myc, PI3K/Akt and mTOR/p70S6K/4E-BP1 and the collaboration with down-regulation of Mcl-1 and survivin may explain KUD983-induced anti-HRPC mechanism.
Collapse
Affiliation(s)
- Mei-Ling Chan
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Chun Yu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jui-Ling Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wohn-Jenn Leu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - She-Hung Chan
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shih-Ping Liu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Polina M Ivantcova
- Department of Medicinal Chemistry, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Özdemir Dogan
- Department of Chemistry, Middle East Technical University, Ankara, Turkey
| | - Stefan Bräse
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Konstantin V Kudryavtsev
- Department of Medicinal Chemistry, Faculty of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation.,Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow region, Russian Federation
| | - Jih-Hwa Guh
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
12
|
Qi Y, Hu Y, Yang H, Zhuang R, Hou Y, Tong H, Feng Y, Huang Y, Jiang Q, Ji Q, Gu Q, Zhang Z, Tang X, Lu W, Zhou Y. Establishing a patient-derived xenograft model of human myxoid and round-cell liposarcoma. Oncotarget 2017; 8:54320-54330. [PMID: 28903344 PMCID: PMC5589583 DOI: 10.18632/oncotarget.17352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 04/10/2017] [Indexed: 12/21/2022] Open
Abstract
Myxoid and round cell liposarcoma (MRCL) is a common type of soft tissue sarcoma. The lack of patient-derived tumor xenograft models that are highly consistent with human tumors has limited the drug experiments for this disease. Hence, we aimed to develop and validate a patient-derived tumor xenograft model of MRCL. A tumor sample from a patient with MRCL was implanted subcutaneously in an immunodeficient mouse shortly after resection to establish a patient-derived tumor xenograft model. After the tumor grew, it was resected and divided into several pieces for re-implantation and tumor passage. After passage 1, 3, and 5 (i.e. P1, P3, and P5, respectively), tumor morphology and the presence of the FUS-DDIT3 gene fusion were consistent with those of the original patient tumor. Short tandem repeat analysis demonstrated consistency from P1 to P5. Whole exome sequencing also showed that P5 tumors harbored many of the same gene mutations present in the original patient tumor, one of which was a PIK3CA mutation. PF-04691502 significantly inhibited tumor growth in P5 models (tumor volumes of 492.62 ± 652.80 vs 3303.81 ± 1480.79 mm3, P < 0.001, in treated vs control tumors, respectively) after 29 days of treatment. In conclusion, we have successfully established the first patient-derived xenograft model of MRCL. In addition to surgery, PI3K/mTOR inhibitors could potentially be used for the treatment of PIK3CA-positive MRCLs.
Collapse
Affiliation(s)
- Yiming Qi
- Departments of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Hu
- Departments of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hua Yang
- Departments of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongyuan Zhuang
- Departments of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingyong Hou
- Departments of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hanxing Tong
- Departments of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Feng
- Departments of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Huang
- Endoscopy Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Quan Jiang
- Departments of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qunsheng Ji
- Oncology BU, Research Service Division, WuXi AppTec, Shanghai, China
| | - Qingyang Gu
- Oncology BU, Research Service Division, WuXi AppTec, Shanghai, China
| | - Zhixiang Zhang
- Oncology BU, Research Service Division, WuXi AppTec, Shanghai, China
| | - Xuzhen Tang
- Oncology BU, Research Service Division, WuXi AppTec, Shanghai, China
| | - Weiqi Lu
- Departments of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuhong Zhou
- Departments of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Schinner C, Vielmuth F, Rötzer V, Hiermaier M, Radeva MY, Co TK, Hartlieb E, Schmidt A, Imhof A, Messoudi A, Horn A, Schlipp A, Spindler V, Waschke J. Adrenergic Signaling Strengthens Cardiac Myocyte Cohesion. Circ Res 2017; 120:1305-1317. [PMID: 28289018 DOI: 10.1161/circresaha.116.309631] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 02/22/2017] [Accepted: 03/10/2017] [Indexed: 01/08/2023]
Abstract
RATIONALE The sympathetic nervous system is a major mediator of heart function. Intercalated discs composed of desmosomes, adherens junctions, and gap junctions provide the structural backbone for coordinated contraction of cardiac myocytes. OBJECTIVE Gap junctions dynamically remodel to adapt to sympathetic signaling. However, it is unknown whether such rapid adaption also occurs for the adhesive function provided by desmosomes and adherens junctions. METHODS AND RESULTS Atomic force microscopy revealed that β-adrenergic signaling enhances both the number of desmoglein 2-specific interactions along cell junctions and the mean desmoglein 2-mediated binding forces, whereas N-cadherin-mediated interactions were not affected. This was accompanied by increased cell cohesion in cardiac myocyte cultures and murine heart slices. Enhanced desmoglein 2-positive contacts and increased junction length as revealed by immunofluorescence and electron microscopy reflected cAMP-induced reorganization of intercellular contacts. The mechanism underlying cAMP-mediated strengthening of desmoglein 2 binding was dependent on expression of the intercalated disc plaque protein plakoglobin (Pg) and direct phosphorylation at S665 by protein kinase A: Pg deficiency as well as overexpression of the phospho-deficient Pg-mutant S665A abrogated both cAMP-mediated junctional remodeling and increase of cohesion. Moreover, Pg knockout hearts failed to functionally adapt to adrenergic stimulation. CONCLUSIONS Taken together, we provide first evidence for positive adhesiotropy as a new cardiac function of sympathetic signaling. Positive adhesiotropy is dependent on Pg phosphorylation at S665 by protein kinase A. This mechanism may be of high medical relevance because loss of junctional Pg is a hallmark of arrhythmogenic cardiomyopathy.
Collapse
Affiliation(s)
- Camilla Schinner
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Franziska Vielmuth
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Vera Rötzer
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Matthias Hiermaier
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Mariya Y Radeva
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Thu Kim Co
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Eva Hartlieb
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Andreas Schmidt
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Axel Imhof
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Ahmed Messoudi
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Anja Horn
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Angela Schlipp
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Volker Spindler
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Jens Waschke
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany.
| |
Collapse
|
14
|
Das TP, Suman S, Papu John AMS, Pal D, Edwards A, Alatassi H, Ankem MK, Damodaran C. Activation of AKT negatively regulates the pro-apoptotic function of death-associated protein kinase 3 (DAPK3) in prostate cancer. Cancer Lett 2016; 377:134-139. [PMID: 27126362 PMCID: PMC4884664 DOI: 10.1016/j.canlet.2016.04.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/14/2016] [Accepted: 04/17/2016] [Indexed: 01/10/2023]
Abstract
The activation of AKT governs many signaling pathways and promotes cell growth and inhibits apoptosis in human malignancies including prostate cancer (CaP). Here, we investigated the molecular association between AKT activation and the function of death-associated protein kinase 3 (DAPK3) in CaP. An inverse correlation of pAKT and DAPK3 expression was seen in a panel of CaP cell lines. Inhibition of AKT by wortmannin/LY294002 or overexpression of DAPK3 reverts the proliferative function of AKT in CaP cells. On the other hand, ectopic expression of AKT inhibited DAPK3 function and induced proliferation of CaP cells. In addition, AKT over-expressed tumors exhibit aggressive growth when compared to control vector in xenograft models. The immunohistochemistry results revealed a down-regulation of DAPK3 expression in AKT over-expressed tumors as compared to control tumors. Finally, we examined the expression pattern of AKT and DAPK3 in human CaP specimens - the expected gradual increase and nuclear localization of pAKT was seen in higher Gleason score samples versus benign hyperplasia (BPH). On the contrary, reduced expression of DAPK3 was seen in higher Gleason stages versus BPH. This suggests that inhibition of DAPK3 may be a contributing factor to the carcinogenesis of the prostate. Understanding the mechanism by which AKT negatively regulates DAPK3 function may suggest whether DAPK3 can be a therapeutic target for CaP.
Collapse
Affiliation(s)
- Trinath P Das
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Suman Suman
- Department of Urology, University of Louisville, Louisville, KY, USA
| | | | - Deeksha Pal
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Angelena Edwards
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Houda Alatassi
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
15
|
Ahearn TU, Tchrakian N, Wilson KM, Lis R, Nuttall E, Sesso HD, Loda M, Giovannucci E, Mucci LA, Finn S, Shui IM. Calcium-Sensing Receptor Tumor Expression and Lethal Prostate Cancer Progression. J Clin Endocrinol Metab 2016; 101:2520-7. [PMID: 27115058 PMCID: PMC4891799 DOI: 10.1210/jc.2016-1082] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CONTEXT Prostate cancer metastases preferentially target bone, and the calcium-sensing receptor (CaSR) may play a role in promoting this metastatic progression. OBJECTIVE We evaluated the association of prostate tumor CaSR expression with lethal prostate cancer. DESIGN A validated CaSR immunohistochemistry assay was performed on tumor tissue microarrays. Vitamin D receptor (VDR) expression and phosphatase and tensin homolog tumor status were previously assessed in a subset of cases by immunohistochemistry. Cox proportional hazards models adjusting for age and body mass index at diagnosis, Gleason grade, and pathological tumor node metastasis stage were used to estimate hazard ratios (HR) and 95% confidence intervals (CI) for the association of CaSR expression with lethal prostate cancer. SETTING The investigation was conducted in the Health Professionals Follow-up Study and Physicians' Health Study. PARTICIPANTS We studied 1241 incident prostate cancer cases diagnosed between 1983 and 2009. MAIN OUTCOME Participants were followed up or cancer-specific mortality or development of metastatic disease. RESULTS On average, men were followed up 13.6 years, during which there were 83 lethal events. High CaSR expression was associated with lethal prostate cancer independent of clinical and pathological variables (HR 2.0; 95% CI 1.2-3.3). Additionally, there was evidence of effect modification by VDR expression; CaSR was associated with lethal progression among men with low tumor VDR expression (HR 3.2; 95% CI 1.4-7.3) but not in cases with high tumor VDR expression (HR 0.8; 95% CI 0.2-3.0). CONCLUSIONS Tumor CaSR expression is associated with an increased risk of lethal prostate cancer, particularly in tumors with low VDR expression. These results support further investigating the mechanism linking CaSR with metastases.
Collapse
Affiliation(s)
- Thomas U Ahearn
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Nairi Tchrakian
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Kathryn M Wilson
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Rosina Lis
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Elizabeth Nuttall
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Howard D Sesso
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Massimo Loda
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Edward Giovannucci
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Lorelei A Mucci
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Stephen Finn
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Irene M Shui
- Departments of Epidemiology (T.U.A., K.M.W., E.N., H.D.S., E.G., L.A.M., I.M.S.) and Department of Nutrition (E.G.), Harvard T. H. Chan School of Public Health, Department of Medical Oncology (R.L., M.L.), Dana-Farber Cancer Institute, Divisions of Preventive Medicine (H.D.S.), and Channing Division of Network Medicine (K.M.W., E.G., L.A.M.), Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115; Department of Histopathology Research (N.T., S.F.), Trinity College, Dublin 8, Ireland; and Public Health Sciences Division (I.M.S.), Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| |
Collapse
|