1
|
Jin R, Forbes CM, Miller NL, Lafin J, Strand DW, Case T, Cates JM, Liu Q, Ramirez-Solano M, Mohler JL, Matusik RJ. Transcriptomic analysis of benign prostatic hyperplasia identifies critical pathways in prostatic overgrowth and 5-alpha reductase inhibitor resistance. Prostate 2024; 84:441-459. [PMID: 38168866 DOI: 10.1002/pros.24661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/06/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND The medical therapy of prostatic symptoms (MTOPS) trial randomized men with symptoms of benign prostatic hyperplasia (BPH) and followed response of treatment with a 5α-reductase inhibitor (5ARI), an alpha-adrenergic receptor antagonist (α-blocker), the combination of 5ARI and α-blocker or no medical therapy (none). Medical therapy reduced risk of clinical progression by 66% but the reasons for nonresponse or loss of therapeutic response in some patients remains unresolved. Our previous work showed that prostatic glucocorticoid levels are increased in 5ARI-treated patients and that glucocorticoids can increased branching of prostate epithelia in vitro. To understand the transcriptomic changes associated with 5ARI treatment, we performed bulk RNA sequencing of BPH and control samples from patients who received 5ARI versus those that did not. Deconvolution analysis was performed to estimate cellular composition. Bulk RNA sequencing was also performed on control versus glucocorticoid-treated prostate epithelia in 3D culture to determine underlying transcriptomic changes associated with branching morphogenesis. METHOD Surgical BPH (S-BPH) tissue was defined as benign prostatic tissue collected from the transition zone (TZ) of patients who failed medical therapy while control tissue termed Incidental BPH (I-BPH) was obtained from the TZ of men undergoing radical prostatectomy for low-volume/grade prostatic adenocarcinoma confined to the peripheral zone. S-BPH patients were divided into four subgroups: men on no medical therapy (none: n = 7), α-blocker alone (n = 10), 5ARI alone (n = 6) or combination therapy (α-blocker and 5ARI: n = 7). Control I-BPH tissue was from men on no medical therapy (none: n = 8) or on α-blocker (n = 6). A human prostatic cell line in 3D culture that buds and branches was used to identify genes involved in early prostatic growth. Snap-frozen prostatic tissue taken at the time of surgery and 3D organoids were used for RNA-seq analysis. Bulk RNAseq data were deconvoluted using CIBERSORTx. Differentially expressed genes (DEG) that were statistically significant among S-BPH, I-BPH, and during budding and branching of organoids were used for pathway analysis. RESULTS Transcriptomic analysis between S-BPH (n = 30) and I-BPH (n = 14) using a twofold cutoff (p < 0.05) identified 377 DEG (termed BPH377) and a cutoff < 0.05 identified 3377 DEG (termed BPH3377). Within the S-BPH, the subgroups none and α-blocker were compared to patients on 5ARI to reveal 361 DEG (termed 5ARI361) that were significantly changed. Deconvolution analysis of bulk RNA seq data with a human prostate single cell data set demonstrated increased levels of mast cells, NK cells, interstitial fibroblasts, and prostate luminal cells in S-BPH versus I-BPH. Glucocorticoid (GC)-induced budding and branching of benign prostatic cells in 3D culture was compared to control organoids to identify early events in prostatic morphogenesis. GC induced 369 DEG (termed GC359) in 3D culture. STRING analysis divided the large datasets into 20-80 genes centered around a hub. In general, biological processes induced in BPH supported growth and differentiation such as chromatin modification and DNA repair, transcription, cytoskeleton, mitochondrial electron transport, ubiquitination, protein folding, and cholesterol synthesis. Identified signaling pathways were pooled to create a list of DEG that fell into seven hubs/clusters. The hub gene centrality was used to name the network including AP-1, interleukin (IL)-6, NOTCH1 and NOTCH3, NEO1, IL-13, and HDAC/KDM. All hubs showed connections to inflammation, chromatin structure, and development. The same approach was applied to 5ARI361 giving multiple networks, but the EGF and sonic hedgehog (SHH) hub was of particular interest as a developmental pathway. The BPH3377, 5ARI363, and GC359 lists were compared and 67 significantly changed DEG were identified. Common genes to the 3D culture included an IL-6 hub that connected to genes identified in BPH hubs that defined AP1, IL-6, NOTCH, NEO1, IL-13, and HDAC/KDM. CONCLUSIONS Reduction analysis of BPH and 3D organoid culture uncovered networks previously identified in prostatic development as being reinitiated in BPH. Identification of these pathways provides insight into the failure of medical therapy for BPH and new therapeutic targets for BPH/LUTS.
Collapse
Affiliation(s)
- Renjie Jin
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Connor M Forbes
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Urology Department, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicole L Miller
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John Lafin
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Douglas W Strand
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas Case
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin M Cates
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marisol Ramirez-Solano
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James L Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Robert J Matusik
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
2
|
Jin R, Forbes C, Miller NL, Strand D, Case T, Cates JM, Kim HYH, Wages P, Porter NA, Mantione KM, Burke S, Mohler JL, Matusik RJ. Glucocorticoids are induced while dihydrotestosterone levels are suppressed in 5-alpha reductase inhibitor treated human benign prostate hyperplasia patients. Prostate 2022; 82:1378-1388. [PMID: 35821619 PMCID: PMC9427722 DOI: 10.1002/pros.24410] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/10/2022] [Accepted: 06/24/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND The development of benign prostatic hyperplasia (BPH) and medication-refractory lower urinary tract symptoms (LUTS) remain poorly understood. This study attempted to characterize the pathways associated with failure of medical therapy for BPH/LUTS. METHODS Transitional zone tissue levels of cholesterol and steroids were measured in patients who failed medical therapy for BPH/LUTS and controls. Prostatic gene expression was measured using qPCR and BPH cells were used in organoid culture to study prostatic branching. RESULTS BPH patients on 5-α-reductase inhibitor (5ARI) showed low levels of tissue dihydrotestosterone (DHT), increased levels of steroid 5-α-reductase type II (SRD5A2), and diminished levels of androgen receptor (AR) target genes, prostate-specific antigen (PSA), and transmembrane serine protease 2 (TMPRSS2). 5ARI raised prostatic tissue levels of glucocorticoids (GC), whereas alpha-adrenergic receptor antagonists (α-blockers) did not. Nuclear localization of GR in prostatic epithelium and stroma appeared in all patient samples. Treatment of four BPH organoid cell lines with dexamethasone, a synthetic GC, resulted in budding and branching. CONCLUSIONS After failure of medical therapy for BPH/LUTS, 5ARI therapy continued to inhibit androgenesis but a 5ARI-induced pathway increased tissue levels of GC not seen in patients on α-blockers. GC stimulation of organoids indicated that the GC receptors are a trigger for controlling growth of prostate glands. A 5ARI-induced pathway revealed GC activation can serve as a master regulator of prostatic branching and growth.
Collapse
Affiliation(s)
- Renjie Jin
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Connor Forbes
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nicole L. Miller
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Douglas Strand
- Department of Urology, University of Texas, Southwestern, Dallas, Texas, USA
| | - Thomas Case
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin M. Cates
- Department of Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hye-Young H. Kim
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Phillip Wages
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Ned A. Porter
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Krystin M. Mantione
- Bioanalytics, Metabolomics, and Pharmacokinetics Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Sarah Burke
- Bioanalytics, Metabolomics, and Pharmacokinetics Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - James L. Mohler
- Bioanalytics, Metabolomics, and Pharmacokinetics Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Robert J. Matusik
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
3
|
CD24 Expression Dampens the Basal Antiviral State in Human Neuroblastoma Cells and Enhances Permissivity to Zika Virus Infection. Viruses 2022; 14:v14081735. [PMID: 36016357 PMCID: PMC9416398 DOI: 10.3390/v14081735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
Zika virus (ZIKV) exhibits distinct selectivity for infection of various cells and tissues, but how host cellular factors modulate varying permissivity remains largely unknown. Previous studies showed that the neuroblastoma cell line SK-N-AS (expressing low levels of cellular protein CD24) was highly restricted for ZIKV infection, and that this restriction was relieved by ectopic expression of CD24. We tested the hypothesis that CD24 expression allowed ZIKV replication by suppression of the antiviral response. SK-N-AS cells expressing an empty vector (termed CD24-low cells) showed elevated basal levels of phosphorylated STAT1, IRF-1, IKKE, and NFκB. In response to exogenously added type I interferon (IFN-I), CD24-low cells had higher-level induction of antiviral genes and activity against two IFN-I-sensitive viruses (VSV and PIV5-P/V) compared to SK-N-AS cells with ectopic CD24 expression (termed CD24-high cells). Media-transfer experiments showed that the inherent antiviral state of CD24-low cells was not dependent on a secreted factor such as IFN-I. Transcriptomics analysis revealed that CD24 expression decreased expression of genes involved in intracellular antiviral pathways, including IFN-I, NFκB, and Ras. Our findings that CD24 expression in neuroblastoma cells represses intracellular antiviral pathways support the proposal that CD24 may represent a novel biomarker in cancer cells for susceptibility to oncolytic viruses.
Collapse
|
4
|
Differential In Vitro Growth and Cell Killing of Cancer versus Benign Prostate Cells by Oncolytic Parainfluenza Virus. Pathogens 2022; 11:pathogens11050493. [PMID: 35631014 PMCID: PMC9147676 DOI: 10.3390/pathogens11050493] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 12/24/2022] Open
Abstract
The development of effective oncolytic viruses will require understanding the differences in virus replication and killing between normal and cancer cells. Here, we have evaluated infections of metastatic cancer (22Rv1) and benign non-tumorigenic (BPH-1) prostate cell lines with a mutant parainfluenza virus 5 (P/V/F) encoding a defective V protein and a hyperfusogenic F protein. Under low multiplicity of infection (MOI), the P/V/F mutant efficiently spread in 22Rv1 cells but was restricted in BPH-1 cells due to type-I interferon (IFN-I) responses. In mixed co-cultures, the P/V/F mutant showed specificity towards and spread within the 22Rv1 cells versus BPH-1 cells. Under high MOI conditions, both BPH-1 and 22Rv1 cells showed efficient infection by the P/V/F mutant. However, compared to BPH-1 cells, the 22Rv1 cancer cells showed increased cytopathic effect, higher induction of caspase-8 and -9, and extensive syncytia formation. In 22Rv1 spheroid cultures, P/V/F infection was less efficient compared to monolayers, but the virus was able to spread through spheroids and induce death. These data indicate that IFN-I sensitivity is a major determinant of specificity of P/V/F spread through populations of cancer versus benign cells, and additionally, differences in activation of apoptotic pathways and syncytia formation can contribute to differential outcomes in cancer versus benign cells.
Collapse
|
5
|
Jin R, Strand DW, Forbes CM, Case T, Cates JM, Liu Q, Ramirez-Solano M, Milne GL, Sanchez S, Wang ZY, Bjorling DE, Miller NL, Matusik RJ. The prostaglandin pathway is activated in patients who fail medical therapy for benign prostatic hyperplasia with lower urinary tract symptoms. Prostate 2021; 81:944-955. [PMID: 34288015 PMCID: PMC8750893 DOI: 10.1002/pros.24190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/16/2021] [Accepted: 06/29/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Little is known about how benign prostatic hyperplasia (BPH) develops and why patients respond differently to medical therapy designed to reduce lower urinary tract symptoms (LUTS). The Medical Therapy of Prostatic Symptoms (MTOPS) trial randomized men with symptoms of BPH and followed response to medical therapy for up to 6 years. Treatment with a 5α-reductase inhibitor (5ARI) or an alpha-adrenergic receptor antagonist (α-blocker) reduced the risk of clinical progression, while men treated with combination therapy showed a 66% decrease in risk of progressive disease. However, medical therapies for BPH/LUTS are not effective in many patients. The reasons for nonresponse or loss of therapeutic response in the remaining patients over time are unknown. A better understanding of why patients fail to respond to medical therapy may have a major impact on developing new approaches for the medical treatment of BPH/LUTS. Prostaglandins (PG) act on G-protein-coupled receptors (GPCRs), where PGE2 and PGF2 elicit smooth muscle contraction. Therefore, we measured PG levels in the prostate tissue of BPH/LUTS patients to assess the possibility that this signaling pathway might explain the failure of medical therapy in BPH/LUTS patients. METHOD Surgical BPH (S-BPH) was defined as benign prostatic tissue collected from the transition zone (TZ) of patients who failed medical therapy and underwent surgical intervention to relieve LUTS. Control tissue was termed Incidental BPH (I-BPH). I-BPH was TZ obtained from men undergoing radical prostatectomy for low-volume, low-grade prostatic adenocarcinoma (PCa, Gleason score ≤ 7) confined to the peripheral zone. All TZ tissue was confirmed to be cancer-free. S-BPH patients divided into four subgroups: patients on α-blockers alone, 5ARI alone, combination therapy (α-blockers plus 5ARI), or no medical therapy (none) before surgical resection. I-BPH tissue was subgrouped by prior therapy (either on α-blockers or without prior medical therapy before prostatectomy). We measured prostatic tissue levels of prostaglandins (PGF2α , PGI2 , PGE2 , PGD2 , and TxA2 ), quantitative polymerase chain reaction levels of mRNAs encoding enzymes within the PG synthesis pathway, cellular distribution of COX1 (PTGS1) and COX2 (PTGS2), and tested the ability of PGs to contract bladder smooth muscle in an in vitro assay. RESULTS All PGs were significantly elevated in TZ tissues from S-BPH patients (n = 36) compared to I-BPH patients (n = 15), regardless of the treatment subgroups. In S-BPH versus I-BPH, mRNA for PG synthetic enzymes COX1 and COX2 were significantly elevated. In addition, mRNA for enzymes that convert the precursor PGH2 to metabolite PGs were variable: PTGIS (which generates PGI2 ) and PTGDS (PGD2 ) were significantly elevated; nonsignificant increases were observed for PTGES (PGE2 ), AKR1C3 (PGF2α ), and TBxAS1 (TxA2 ). Within the I-BPH group, men responding to α-blockers for symptoms of BPH but requiring prostatectomy for PCa did not show elevated levels of COX1, COX2, or PGs. By immunohistochemistry, COX1 was predominantly observed in the prostatic stroma while COX2 was present in scattered luminal cells of isolated prostatic glands in S-BPH. PGE2 and PGF2α induced contraction of bladder smooth muscle in an in vitro assay. Furthermore, using the smooth muscle assay, we demonstrated that α-blockers that inhibit alpha-adrenergic receptors do not appear to inhibit PG stimulation of GPCRs in bladder muscle. Only patients who required surgery to relieve BPH/LUTS symptoms showed significantly increased tissue levels of PGs and the PG synthetic enzymes. CONCLUSIONS Treatment of BPH/LUTS by inhibition of alpha-adrenergic receptors with pharmaceutical α-blockers or inhibiting androgenesis with 5ARI may fail because of elevated paracrine signaling by prostatic PGs that can cause smooth muscle contraction. In contrast to patients who fail medical therapy for BPH/LUTS, control I-BPH patients do not show the same evidence of elevated PG pathway signaling. Elevation of the PG pathway may explain, in part, why the risk of clinical progression in the MTOPS study was only reduced by 34% with α-blocker treatment.
Collapse
Affiliation(s)
- Renjie Jin
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Connor M. Forbes
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas Case
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin M.M. Cates
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qi Liu
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marisol Ramirez-Solano
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ginger L. Milne
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephanie Sanchez
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zunyi Y. Wang
- School of Veterinary Medicine, University of Wisconsin, Madison, WI
| | - Dale E. Bjorling
- School of Veterinary Medicine, University of Wisconsin, Madison, WI
| | - Nicole L. Miller
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert J. Matusik
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
- Corresponding author Robert J. Matusik, Ph.D., Department of Urology, A1302 MCN, Vanderbilt University Medical Center, Nashville, TN 37232,
| |
Collapse
|
6
|
Thomas S, Dunn CD, Campbell LJ, Strand DW, Vezina CM, Bjorling DE, Penniston KL, Li L, Ricke WA, Goldberg TL. A multi-omic investigation of male lower urinary tract symptoms: Potential role for JC virus. PLoS One 2021; 16:e0246266. [PMID: 33630889 PMCID: PMC7906371 DOI: 10.1371/journal.pone.0246266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 01/15/2021] [Indexed: 12/22/2022] Open
Abstract
Male lower urinary tract symptoms (LUTS) comprise a common syndrome of aging that negatively impacts quality of life. The etiology of LUTS is multifactorial, involving benign prostatic hyperplasia, smooth muscle and neurologic dysfunction, inflammation, sexually transmitted infections, fibrosis, and potentially dysbiosis, but this aspect remains poorly explored. We investigated whether the presence of infectious agents in urine might be associated with LUTS by combining next-generation DNA sequencing for virus discovery, microbiome analysis for characterization of bacterial communities, and mass spectrometry-based metabolomics. In urine from 29 LUTS cases and 9 controls from Wisconsin, we found a statistically significant association between a diagnosis of LUTS and the presence of JC virus (JCV), a common neurotropic human polyomavirus (Polyomaviridae, Betapolyomavirus) linked to severe neurologic disease in rare cases. This association (based on metagenomics) was not borne out when specific polymerase chain reaction (PCR) testing was applied to this set of samples, likely due to the greater sensitivity of PCR. Interestingly, urine metabolomics analysis identified dysregulation of metabolites associated with key LUTS processes. Microbiome analysis found no evidence of microbial community dysbiosis in LUTS cases, but JCV-positive samples contained more Anaerococcus species, which are involved in polymicrobial infections of the urinary tract. Neither age nor body mass index were significantly associated with the presence of urinary JCV-in the initial group or in an additional, regionally distinct group. These data provide preliminary support the hypothesis that viruses such as JCV may play a role in the development or progression of LUTS, together with other infectious agents and host metabolic responses.
Collapse
Affiliation(s)
- Samuel Thomas
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Christopher D. Dunn
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Lewis J. Campbell
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Douglas W. Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Chad M. Vezina
- George M. O’Brien Center of Research Excellence, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Dale E. Bjorling
- George M. O’Brien Center of Research Excellence, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kristina L. Penniston
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Lingjun Li
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - William A. Ricke
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- George M. O’Brien Center of Research Excellence, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Tony L. Goldberg
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- UW-Madison Global Health Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
7
|
Silva JAF, Calmasini F, Siqueira-Berti A, Moraes-Vieira PMM, Quintar A, Carvalho HF. Prostate immunology: A challenging puzzle. J Reprod Immunol 2020; 142:103190. [PMID: 32853844 DOI: 10.1016/j.jri.2020.103190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/30/2020] [Accepted: 08/11/2020] [Indexed: 12/15/2022]
Abstract
Mucosal immunity defines the relationship of surfaces in contact with the environment and integrates diverse tissues such as epidermis, gum, nose, gut, uterus and prostate with the immune system. Although considered part of a system, each mucosa presents specific immune features beyond the barrier and secretory functions. Information regarding the mucosal immunology of the male reproductive tract and the prostate gland in particular is scarce. In this review, we approach the prostate as an epithelial barrier and as part of the mucosal immune system. Finally, we also raise a series of questions that will improve the understanding of this gland, its role in reproduction and its sensitivity/resistance to disease.
Collapse
Affiliation(s)
- Juliete Aparecida F Silva
- Department of Structural and Functional Biology, State University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Fabiano Calmasini
- Department of Structural and Functional Biology, State University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Aline Siqueira-Berti
- Department of Structural and Functional Biology, State University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Pedro M M Moraes-Vieira
- Department of Genetics, Evolution, Microbiology and Immunology, State University of Campinas, UNICAMP, Campinas, SP, Brazil
| | - Amado Quintar
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, State University of Campinas - UNICAMP, Campinas, SP, Brazil; National Institute of Science and Technology of Photonics Applied to Cell Biology - INFABiC, Campinas, SP, Brazil.
| |
Collapse
|
8
|
Fox CR, Parks GD. Histone Deacetylase Inhibitors Enhance Cell Killing and Block Interferon-Beta Synthesis Elicited by Infection with an Oncolytic Parainfluenza Virus. Viruses 2019; 11:E431. [PMID: 31083335 PMCID: PMC6563284 DOI: 10.3390/v11050431] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/28/2022] Open
Abstract
Previous results have shown that infection with the cytoplasmic-replicating parainfluenza virus 5 mutant P/V-CPI- sensitizes cells to DNA damaging agents, resulting in the enhanced killing of airway cancer cells. Here, we have tested the hypothesis that histone deacetylase (HDAC) inhibitors can also act with P/V-CPI- infection to enhance cancer cell killing. Using human small cell lung cancer and laryngeal cancer cell lines, 10 HDAC inhibitors were tested for their effect on viability of P/V-CPI- infected cells. HDAC inhibitors such as scriptaid enhanced caspase-3/7, -8 and -9 activity induced by P/V-CPI- and overall cell toxicity. Scriptaid-mediated enhanced killing was eliminated in lung cancer cells that were engineered to express a protein which sequesters double stranded RNA. Scriptaid also enhanced cancer cell killing by two other negative strand RNA viruses - the La Crosse virus and vesicular stomatitis virus. Scriptaid treatment enhanced the spread of the P/V-CPI- virus through a population of cancer cells, and suppressed interferon-beta induction through blocking phosphorylation and nuclear translocation of Interferon Regulatory Factor 3 (IRF-3). Taken together, these data support a role for combinations of a cytoplasmic-replicating RNA virus such as the P/V-CPI- mutant along with chemotherapeutic agents.
Collapse
Affiliation(s)
- Candace R Fox
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.
| | - Griffith D Parks
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.
| |
Collapse
|
9
|
Joseph DB, Strand DW, Vezina CM. DNA methylation in development and disease: an overview for prostate researchers. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2018; 6:197-218. [PMID: 30697577 PMCID: PMC6334199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/14/2018] [Indexed: 06/09/2023]
Abstract
Epigenetic mechanisms including DNA methylation are critical regulators of organismal development and tissue homeostasis. DNA methylation is the transfer of methyl groups to cytosines, which adds an additional layer of complexity to the genome. DNA methylation marks are recognized by the cellular machinery to regulate transcription. Disruption of DNA methylation with aging or exposure to environmental toxins can change susceptibility to disease or trigger processes that lead to disease. In this review, we provide an overview of the DNA methylation machinery. More specifically, we describe DNA methylation in the context of prostate development, prostate cancer, and benign prostatic hyperplasia (BPH) as well as the impact of dietary and environmental factors on DNA methylation in the prostate.
Collapse
Affiliation(s)
- Diya B Joseph
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI 53706, USA
| | - Douglas W Strand
- Department of Urology, UT Southwestern Medical CenterDallas, TX 75390, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin-MadisonMadison, WI 53706, USA
| |
Collapse
|
10
|
Austin DC, Strand DW, Love HL, Franco OE, Jang A, Grabowska MM, Miller NL, Hameed O, Clark PE, Fowke JH, Matusik RJ, Jin RJ, Hayward SW. NF-κB and androgen receptor variant expression correlate with human BPH progression. Prostate 2016; 76:491-511. [PMID: 26709083 PMCID: PMC4763342 DOI: 10.1002/pros.23140] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is a common, chronic progressive disease. Inflammation is associated with prostatic enlargement and resistance to 5α-reductase inhibitor (5ARI) therapy. Activation of the nuclear factor-kappa B (NF-κB) pathway is linked to both inflammation and ligand-independent prostate cancer progression. METHODS NF-κB activation and androgen receptor variant (AR-V) expression were quantified in transition zone tissue samples from patients with a wide range of AUASS from incidental BPH in patients treated for low grade, localized peripheral zone prostate cancer to advanced disease requiring surgical intervention. To further investigate these pathways, human prostatic stromal and epithelial cell lines were transduced with constitutively active or kinase dead forms of IKK2 to regulate canonical NF-κB activity. The effects on AR full length (AR-FL) and androgen-independent AR-V expression as well as cellular growth and differentiation were assessed. RESULTS Canonical NF-κB signaling was found to be upregulated in late versus early stage BPH, and to be strongly associated with non-insulin dependent diabetes mellitus. Elevated expression of AR-variant 7 (AR-V7), but not other AR variants, was found in advanced BPH samples. Expression of AR-V7 significantly correlated with the patient AUASS and TRUS volume. Forced activation of canonical NF-κB in human prostatic epithelial and stromal cells resulted in elevated expression of both AR-FL and AR-V7, with concomitant ligand-independent activation of AR reporters. Activation of NF-κB and over expression of AR-V7 in human prostatic epithelial cells maintained cell viability in the face of 5ARI treatment. CONCLUSION Activation of NF-κB and AR-V7 in the prostate is associated with increased disease severity. AR-V7 expression is inducible in human prostate cells by forced activation of NF-κB resulting in resistance to 5ARI treatment, suggesting a potential mechanism by which patients may become resistant to 5ARI therapy.
Collapse
Affiliation(s)
- David C Austin
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas W Strand
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Harold L Love
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Omar E Franco
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - Alex Jang
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Magdalena M Grabowska
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nicole L Miller
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Omar Hameed
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Peter E Clark
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jay H Fowke
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert J Matusik
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ren J Jin
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Simon W Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| |
Collapse
|
11
|
Keil KP, Vezina CM. DNA methylation as a dynamic regulator of development and disease processes: spotlight on the prostate. Epigenomics 2015; 7:413-25. [PMID: 26077429 DOI: 10.2217/epi.15.8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prostate development, benign hyperplasia and cancer involve androgen and growth factor signaling as well as stromal-epithelial interactions. We review how DNA methylation influences these and related processes in other organ systems such as how proliferation is restricted to specific cell populations during defined temporal windows, how androgens elicit their actions and how cells establish, maintain and remodel DNA methylation in a time and cell specific fashion. We also discuss mechanisms by which hormones and endocrine disrupting chemicals reprogram DNA methylation in the prostate and elsewhere and examine evidence for a reawakening of developmental epigenetic pathways as drivers of prostate cancer and benign prostate hyperplasia.
Collapse
Affiliation(s)
- Kimberly P Keil
- Comparative Biosciences, University of Wisconsin-Madison, 1656 Linden Dr., Madison, WI 53705, USA
| | - Chad M Vezina
- Comparative Biosciences, University of Wisconsin-Madison, 1656 Linden Dr., Madison, WI 53705, USA
| |
Collapse
|
12
|
Hickling DR, Sun TT, Wu XR. Anatomy and Physiology of the Urinary Tract: Relation to Host Defense and Microbial Infection. Microbiol Spectr 2015; 3:10.1128/microbiolspec.UTI-0016-2012. [PMID: 26350322 PMCID: PMC4566164 DOI: 10.1128/microbiolspec.uti-0016-2012] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Indexed: 02/07/2023] Open
Abstract
The urinary tract exits to a body surface area that is densely populated by a wide range of microbes. Yet, under most normal circumstances, it is typically considered sterile, i.e., devoid of microbes, a stark contrast to the gastrointestinal and upper respiratory tracts where many commensal and pathogenic microbes call home. Not surprisingly, infection of the urinary tract over a healthy person's lifetime is relatively infrequent, occurring once or twice or not at all for most people. For those who do experience an initial infection, the great majority (70% to 80%) thankfully do not go on to suffer from multiple episodes. This is a far cry from the upper respiratory tract infections, which can afflict an otherwise healthy individual countless times. The fact that urinary tract infections are hard to elicit in experimental animals except with inoculum 3-5 orders of magnitude greater than the colony counts that define an acute urinary infection in humans (105 cfu/ml), also speaks to the robustness of the urinary tract defense. How can the urinary tract be so effective in fending off harmful microbes despite its orifice in a close vicinity to that of the microbe-laden gastrointestinal tract? While a complete picture is still evolving, the general consensus is that the anatomical and physiological integrity of the urinary tract is of paramount importance in maintaining a healthy urinary tract. When this integrity is breached, however, the urinary tract can be at a heightened risk or even recurrent episodes of microbial infections. In fact, recurrent urinary tract infections are a significant cause of morbidity and time lost from work and a major challenge to manage clinically. Additionally, infections of the upper urinary tract often require hospitalization and prolonged antibiotic therapy. In this chapter, we provide an overview of the basic anatomy and physiology of the urinary tract with an emphasis on their specific roles in host defense. We also highlight the important structural and functional abnormalities that predispose the urinary tract to microbial infections.
Collapse
Affiliation(s)
- Duane R Hickling
- Division of Urology, Ottawa Hospital Research Institute, The Ottawa Hospital, University of Ottawa, Ottawa, ON K1Y 4E9, Canada
| | - Tung-Tien Sun
- Departments of Cell Biology, Biochemistry and Molecular Pharmacology, Departments of Dermatology and Urology, New York University School of Medicine, New York, NY, 10016
| | - Xue-Ru Wu
- Departments of Urology and Pathology, New York University School of Medicine, New York, NY, 10016
| |
Collapse
|
13
|
|
14
|
Lin-Tsai O, Clark PE, Miller NL, Fowke JH, Hameed O, Hayward SW, Strand DW. Surgical intervention for symptomatic benign prostatic hyperplasia is correlated with expression of the AP-1 transcription factor network. Prostate 2014; 74:669-79. [PMID: 24500928 PMCID: PMC4160824 DOI: 10.1002/pros.22785] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 01/06/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Approximately one-third of patients fail medical treatment for benign prostatic hyperplasia and associated lower urinary tract symptoms (BPH/LUTS) requiring surgical intervention. Our purpose was to establish a molecular characterization for patients undergoing surgical intervention for LUTS to address therapeutic deficiencies. METHODS Clinical, molecular, and histopathological profiles were analyzed in 26 patients undergoing surgery for severe LUTS. Incidental transitional zone nodules were isolated from 37 patients with mild symptoms undergoing radical prostatectomy. Clinical parameters including age, prostate volume, medication, prostate specific antigen, symptom score, body mass index, and incidence of diabetes were collected. Multivariate logistic regression analysis with adjustments for potential confounding variables was used to examine associations between patient clinical characteristics and molecular targets identified through molecular profiling. RESULTS Compared to incidental BPH, progressive symptomatic BPH was associated with increased expression of the activating protein-1 transcription factor/chemokine network. As expected, inverse correlations were drawn between androgen receptor levels and age, as well as between 5α-reductase inhibitor (5ARI) treatment and tissue prostate specific antigen levels; however, a novel association was also drawn between 5ARI treatment and increased c-FOS expression. CONCLUSIONS This study provides molecular evidence that a network of pro-inflammatory activating protein-1 transcription factors and associated chemokines are highly enriched in symptomatic prostate disease, a profile that molecularly categorizes with many other chronic autoimmune diseases. Because 5ARI treatment was associated with increased c-FOS expression, future studies should explore whether increased activating protein-1 proteins are causal factors in the development of symptomatic prostate disease, inflammation or resistance to traditional hormonal therapy.
Collapse
Affiliation(s)
- Opal Lin-Tsai
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Peter E. Clark
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nicole L. Miller
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jay H. Fowke
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Omar Hameed
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Simon W. Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas W. Strand
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Correspondence to: Douglas W. Strand, Department of Urologic Surgery, A-1302 Vanderbilt University Medical Center, Nashville, TN 37232-2765.
| |
Collapse
|
15
|
Kaarbø M, Storm ML, Qu S, Wæhre H, Risberg B, Danielsen HE, Saatcioglu F. TCTP is an androgen-regulated gene implicated in prostate cancer. PLoS One 2013; 8:e69398. [PMID: 23894469 PMCID: PMC3718683 DOI: 10.1371/journal.pone.0069398] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 06/10/2013] [Indexed: 11/19/2022] Open
Abstract
TCTP has been implicated in a plethora of important cellular processes related to cell growth, cell cycle progression, malignant transformation and inhibition of apoptosis. In addition to these intracellular functions, TCTP has extracellular functions and plays an important role in immune cells. TCTP expression was previously shown to be deregulated in prostate cancer, but its function in prostate cancer cells is largely unknown. Here we show that TCTP expression is regulated by androgens in LNCaP prostate cancer cells in vitro as well as human prostate cancer xenografts in vivo. Knockdown of TCTP reduced colony formation and increased apoptosis in LNCaP cells, implicating it as an important factor for prostate cancer cell growth. Global gene expression profiling in TCTP knockdown LNCaP cells showed that several interferon regulated genes are regulated by TCTP, suggesting that it may have a role in regulating immune function in prostate cancer. In addition, recombinant TCTP treatment increased colony formation in LNCaP cells suggesting that secreted TCTP may function as a proliferative factor in prostate cancer. These results suggest that TCTP may have a role in prostate cancer development.
Collapse
Affiliation(s)
- Mari Kaarbø
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Su Qu
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Håkon Wæhre
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
| | - Bjørn Risberg
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
- Division of Pathology, Oslo University Hospital, Oslo, Norway
| | - Håvard E. Danielsen
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
- Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|