1
|
Procházková J, Kahounová Z, Vondráček J, Souček K. Aryl hydrocarbon receptor as a drug target in advanced prostate cancer therapy - obstacles and perspectives. Transcription 2025; 16:47-66. [PMID: 38547312 PMCID: PMC11970783 DOI: 10.1080/21541264.2024.2334106] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2025] Open
Abstract
Aryl hydrocarbon receptor (AhR) is a transcription factor that is primarily known as an intracellular sensor of environmental pollution. After five decades, the list of synthetic and toxic chemicals that activate AhR signaling has been extended to include a number of endogenous compounds produced by various types of cells via their metabolic activity. AhR signaling is active from the very beginning of embryonal development throughout the life cycle and participates in numerous biological processes such as control of cell proliferation and differentiation, metabolism of aromatic compounds of endogenous and exogenous origin, tissue regeneration and stratification, immune system development and polarization, control of stemness potential, and homeostasis maintenance. AhR signaling can be affected by various pharmaceuticals that may help modulate abnormal AhR signaling and drive pathological states. Given their role in immune system development and regulation, AhR antagonistic ligands are attractive candidates for immunotherapy of disease states such as advanced prostate cancer, where an aberrant immune microenvironment contributes to cancer progression and needs to be reeducated. Advanced stages of prostate cancer are therapeutically challenging and characterized by decreased overall survival (OS) due to the metastatic burden. Therefore, this review addresses the role of AhR signaling in the development and progression of prostate cancer and discusses the potential of AhR as a drug target for the treatment of advanced prostate cancer upon entering the phase of drug resistance and failure of first-line androgen deprivation therapy.Abbreviation: ADC: antibody-drug conjugate; ADT: androgen deprivation therapy; AhR: aryl hydrocarbon receptor; AR: androgen receptor; ARE: androgen response element; ARPI: androgen receptor pathway inhibitor; mCRPC: metastatic castration-resistant prostate cancer; DHT: 5a-dihydrotestosterone; FICZ: 6-formylindolo[3,2-b]carbazole; 3-MC: 3-methylcholanthrene; 6-MCDF: 6-methyl-1,3,8-trichlorodibenzofuran; MDSCs: myeloid-derived suppressor cells; PAHs: polycyclic aromatic hydrocarbons; PCa: prostate cancer; TAMs: tumor-associated macrophages; TF: transcription factor; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; TME: tumor microenvironment; TRAMP: transgenic adenocarcinoma of the mouse prostate; TROP2: tumor associated calcium signal transducer 2.
Collapse
Affiliation(s)
- Jiřina Procházková
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Zuzana Kahounová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|
2
|
Lavery TC, Spiegelhoff A, Wang K, Kennedy CL, Ridlon M, Keil Stietz KP. Polychlorinated biphenyl (PCB) exposure in adult female mice can influence bladder contractility. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2023; 11:367-384. [PMID: 37941647 PMCID: PMC10628623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/21/2023] [Indexed: 11/10/2023]
Abstract
Lower urinary tract symptoms (LUTS) greatly reduce quality of life. While LUTS etiology is not completely understood, it is plausible that environmental contaminants could play a role. Polychlorinated biphenyls (PCBs), are a group of persistent environmental toxicants frequently documented in animal and human tissues. PCBs are capable of influencing voiding function in mouse offspring exposed developmentally, however whether PCB exposure during adulthood can also influence voiding dynamics is unknown. Therefore, the purpose of this study was to determine whether PCB exposure in adult female mice can impact voiding function. As part of a larger study to generate developmentally exposed offspring, adult female C57Bl/6J mice were dosed orally with the MARBLES PCB mixture (0.1, 1, or 6 mg/kg/day) or vehicle control beginning two weeks before mating and throughout gestation and lactation (9 weeks). Adult dosed female dams then underwent void spot assay, uroflowmetry, and anesthetized cystometry to assess voiding function. Bladder contractility was assessed in ex vivo bladder bath assays, and bladders were collected for morphology and histology assessments. While voiding behavior endpoints were minimally impacted, alterations to bladder contractility dynamics were more pronounced. Adult female mice dosed with 1 mg/kg/d PCB showed an increase in urine spots 2-3 cm2 in size, increased bladder contractility in response to electrical field stimulation, and decreased bladder wall thickness compared to vehicle control. PCBs also altered contractile response to cholinergic agonist in a dose-dependent manner. Overall, these results indicate that exposure to PCBs in adult female mice is sufficient to produce changes in bladder physiology. These results also highlight the critical role of timing of exposure in influencing voiding function.
Collapse
Affiliation(s)
- Thomas Cm Lavery
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison Madison, Wisconsin 53706, USA
| | - Audrey Spiegelhoff
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison Madison, Wisconsin 53706, USA
| | - Kathy Wang
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison Madison, Wisconsin 53706, USA
| | - Conner L Kennedy
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison Madison, Wisconsin 53706, USA
| | - Monica Ridlon
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison Madison, Wisconsin 53706, USA
| | - Kimberly P Keil Stietz
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison Madison, Wisconsin 53706, USA
| |
Collapse
|
3
|
Spiegelhoff A, Wang K, Ridlon M, Lavery T, Kennedy CL, George S, Stietz KPK. Polychlorinated Biphenyls (PCBs) Impact Prostatic Collagen Density and Bladder Volume in Young Adult Mice Exposed during in Utero and Lactational Development. TOXICS 2023; 11:609. [PMID: 37505574 PMCID: PMC10384510 DOI: 10.3390/toxics11070609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/29/2023]
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic pollutants linked to deleterious health outcomes, including voiding dysfunction in developmentally exposed mice. Changes in prostate volume and/or extracellular matrix composition are associated with voiding dysfunction in men and animal models. Whether PCB-induced changes in voiding function in male mice occur in part via alterations to the prostate or an alternate mechanism is unclear. Therefore, we tested whether developmental exposure to the MARBLES PCB mixture altered prostate morphology in young adult offspring. C57Bl/6J female mice were dosed daily with the MARBLES PCB mixture at 0, 0.1, 1 or 6 mg/kg/d for two weeks prior to mating and through gestation and lactation, offspring were collected at 6 weeks of age. Ventral prostate mass was decreased in the 1 mg/kg/d PCB group compared to other PCB groups. There were no PCB-induced changes in prostate smooth muscle thickness, apoptosis, proliferation, or testes mass. PCBs impacted the prostate extracellular matrix; anterior prostate collagen density was decreased in the 1 mg/kg/d PCB group compared to all other groups. Normalized bladder volume was increased in male and female offspring in the 6 mg/kg/d PCB group compared to control. No change in water consumption, bladder mass or bladder smooth muscle thickness accompanied changes in bladder volume. Urine and serum creatinine concentrations were elevated but only in male mice. Together, these results suggest that developmental exposure to PCBs can influence prostate wet weight and prostate/bladder morphology, but PCBs do not promote prostate enlargement. Whether these changes persist throughout adult life and how they contribute to voiding function in animal models and humans is of future interest.
Collapse
Affiliation(s)
- Audrey Spiegelhoff
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kathy Wang
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Monica Ridlon
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Thomas Lavery
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Conner L Kennedy
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Serena George
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kimberly P Keil Stietz
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
4
|
Wang Z, Spitz R, Vezina C, Hou J, Bjorling DE. Lack of expression of miR-29a/b1 impairs bladder function in male mice. Dis Model Mech 2023; 16:dmm050054. [PMID: 37283037 PMCID: PMC10259841 DOI: 10.1242/dmm.050054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Lower urinary tract symptoms (LUTS) refer to various urological diseases, and incomplete bladder emptying is common among affected patients. The etiology of LUTS is largely unknown, and investigations of LUTS suggest that bladder fibrosis contributes to pathogenesis of LUTS. MicroRNAs (miRNAs) are short (∼22 nucleotides), non-coding RNAs that repress target gene expression by a combination of mRNA degradation and translation inhibition. The miR-29 family is best known for its anti-fibrotic role in various organs. miR-29 was decreased in bladders of patients with outlet obstruction and a rat model of bladder outlet obstruction, suggesting that miR-29 may contribute to impaired bladder function subsequent to tissue fibrosis. We characterized bladder function in male mice lacking expression of Mir29a and Mir29b-1 (miR-29a/b1). Lack of miR-29a/b1 resulted in severe urinary retention, increased voiding duration and reduced flow rate, and these mice failed to void or voided irregularly during anesthetized cytometry. Collagens and elastin were increased in bladders of mice lacking miR-29a/b1. These findings reveal an important role for miR-29 in bladder homeostasis and suggest the therapeutic potential of miR-29 to improve symptoms in patients with LUTS.
Collapse
Affiliation(s)
- Zunyi Wang
- Department of Surgical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Robert Spitz
- Department of Surgical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chad Vezina
- The O'Brien Center for Urologic Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jianghui Hou
- Division of Nephrology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Dale E. Bjorling
- Department of Surgical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
- The O'Brien Center for Urologic Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Urology, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
5
|
From Nucleus to Organs: Insights of Aryl Hydrocarbon Receptor Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms232314919. [PMID: 36499247 PMCID: PMC9738205 DOI: 10.3390/ijms232314919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a markedly established regulator of a plethora of cellular and molecular processes. Its initial role in the detoxification of xenobiotic compounds has been partially overshadowed by its involvement in homeostatic and organ physiology processes. In fact, the discovery of its ability to bind specific target regulatory sequences has allowed for the understanding of how AHR modulates such processes. Thereby, AHR presents functions in transcriptional regulation, chromatin architecture modifications and participation in different key signaling pathways. Interestingly, such fields of influence end up affecting organ and tissue homeostasis, including regenerative response both to endogenous and exogenous stimuli. Therefore, from classical spheres such as canonical transcriptional regulation in embryonic development, cell migration, differentiation or tumor progression to modern approaches in epigenetics, senescence, immune system or microbiome, this review covers all aspects derived from the balance between regulation/deregulation of AHR and its physio-pathological consequences.
Collapse
|
6
|
Kennedy CL, Spiegelhoff A, Lavery T, Wang K, Manuel RSJ, Wang Z, Wildermuth H, Keil Stietz KP. Developmental polychlorinated biphenyl (PCB) exposure alters voiding physiology in young adult male and female mice. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2022; 10:82-97. [PMID: 35528463 PMCID: PMC9077147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/10/2022] [Indexed: 06/14/2023]
Abstract
The impact of developmental exposure to environmental chemicals on lower urinary tract function is not well understood, despite the fact that these chemicals could contribute to etiologically complex lower urinary tract symptoms (LUTS). Polychlorinated biphenyls (PCBs) are environmental toxicants known to be detrimental to the central nervous system, but their impact on voiding function in mouse models is not known. Therefore, we test whether developmental exposure to PCBs is capable of altering voiding physiology in young adult mice. C57Bl/6J female mice received a daily oral dose of the MARBLES PCB mixture for two weeks prior to mating and through gestation and lactation. The mixture mimics the profile of PCBs found in a contemporary population of pregnant women. Voiding function was then tested in young adult offspring using void spot assay, uroflowmetry and anesthetized cystometry. PCB effects were sex and dose dependent. Overall, PCBs led to increases in small size urine spots in both sexes with males producing more drop-like voids and greater peak pressure during a voiding cycle while females displayed decreases in void duration and intervoid interval. Together, these results indicate that developmental exposure to PCBs are capable of altering voiding physiology in young adult mice. Further work to identify the underlying mechanisms driving these changes may help develop more effective preventative or therapeutic strategies for LUTS.
Collapse
Affiliation(s)
- Conner L Kennedy
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI 53706, USA
| | - Audrey Spiegelhoff
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI 53706, USA
| | - Thomas Lavery
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI 53706, USA
| | - Kathy Wang
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI 53706, USA
| | - Robbie SJ Manuel
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI 53706, USA
| | - Zunyi Wang
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI 53706, USA
| | - Hannah Wildermuth
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI 53706, USA
| | - Kimberly P Keil Stietz
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI 53706, USA
| |
Collapse
|
7
|
Male Lower Urinary Tract Dysfunction: An Underrepresented Endpoint in Toxicology Research. TOXICS 2022; 10:toxics10020089. [PMID: 35202275 PMCID: PMC8880407 DOI: 10.3390/toxics10020089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023]
Abstract
Lower urinary tract dysfunction (LUTD) is nearly ubiquitous in men of advancing age and exerts substantial physical, mental, social, and financial costs to society. While a large body of research is focused on the molecular, genetic, and epigenetic underpinnings of the disease, little research has been dedicated to the influence of environmental chemicals on disease initiation, progression, or severity. Despite a few recent studies indicating a potential developmental origin of male LUTD linked to chemical exposures in the womb, it remains a grossly understudied endpoint in toxicology research. Therefore, we direct this review to toxicologists who are considering male LUTD as a new aspect of chemical toxicity studies. We focus on the LUTD disease process in men, as well as in the male mouse as a leading research model. To introduce the disease process, we describe the physiology of the male lower urinary tract and the cellular composition of lower urinary tract tissues. We discuss known and suspected mechanisms of male LUTD and examples of environmental chemicals acting through these mechanisms to contribute to LUTD. We also describe mouse models of LUTD and endpoints to diagnose, characterize, and quantify LUTD in men and mice.
Collapse
|
8
|
Kennedy CL, Spiegelhoff A, Wang K, Lavery T, Nunez A, Manuel R, Hillers-Ziemer L, Arendt LM, Stietz KPK. The Bladder Is a Novel Target of Developmental Polychlorinated Biphenyl Exposure Linked to Increased Inflammatory Cells in the Bladder of Young Mice. TOXICS 2021; 9:toxics9090214. [PMID: 34564365 PMCID: PMC8473463 DOI: 10.3390/toxics9090214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 01/28/2023]
Abstract
Bladder inflammation is associated with several lower urinary tract symptoms that greatly reduce quality of life, yet contributing factors are not completely understood. Environmental chemicals are plausible mediators of inflammatory reactions within the bladder. Here, we examine whether developmental exposure to polychlorinated biphenyls (PCBs) leads to changes in immune cells within the bladder of young mice. Female mice were exposed to an environmentally relevant mixture of PCBs through gestation and lactation, and bladders were collected from offspring at postnatal day (P) 28-31. We identify several dose- and sex-dependent PCB effects in the bladder. The lowest concentration of PCB (0.1 mg/kg/d) increased CD45+ hematolymphoid immune cells in both sexes. While PCBs had no effect on CD79b+ B cells or CD3+ T cells, PCBs (0.1 mg/kg/d) did increase F4/80+ macrophages particularly in female bladder. Collagen density was also examined to determine whether inflammatory events coincide with changes in the stromal extracellular matrix. PCBs (0.1 mg/kg/d) decreased collagen density in female bladder compared to control. PCBs also increased the number of cells undergoing cell division predominantly in male bladder. These results implicate perturbations to the immune system in relation to PCB effects on the bladder. Future study to define the underlying mechanisms could help understand how environmental factors can be risk factors for lower urinary tract symptoms.
Collapse
|
9
|
Turco AE, Oakes SR, Keil Stietz KP, Dunham CL, Joseph DB, Chathurvedula TS, Girardi NM, Schneider AJ, Gawdzik J, Sheftel CM, Wang P, Wang Z, Bjorling DE, Ricke WA, Tang W, Hernandez LL, Keast JR, Bonev AD, Grimes MD, Strand DW, Tykocki NR, Tanguay RL, Peterson RE, Vezina CM. A mechanism linking perinatal 2,3,7,8 tetrachlorodibenzo-p-dioxin exposure to lower urinary tract dysfunction in adulthood. Dis Model Mech 2021; 14:271057. [PMID: 34318329 PMCID: PMC8326766 DOI: 10.1242/dmm.049068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Benign prostatic hyperplasia/lower urinary tract dysfunction (LUTD) affects nearly all men. Symptoms typically present in the fifth or sixth decade and progressively worsen over the remainder of life. Here, we identify a surprising origin of this disease that traces back to the intrauterine environment of the developing male, challenging paradigms about when this disease process begins. We delivered a single dose of a widespread environmental contaminant present in the serum of most Americans [2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD), 1 µg/kg], and representative of a broader class of environmental contaminants, to pregnant mice and observed an increase in the abundance of a neurotrophic factor, artemin, in the developing mouse prostate. Artemin is required for noradrenergic axon recruitment across multiple tissues, and TCDD rapidly increases prostatic noradrenergic axon density in the male fetus. The hyperinnervation persists into adulthood, when it is coupled to autonomic hyperactivity of prostatic smooth muscle and abnormal urinary function, including increased urinary frequency. We offer new evidence that prostate neuroanatomical development is malleable and that intrauterine chemical exposures can permanently reprogram prostate neuromuscular function to cause male LUTD in adulthood. Summary: We describe a new mechanism of benign prostate disease, initiated by fetal chemical exposure, which durably increases prostatic noradrenergic axon density and causes smooth muscle hyperactivity and urinary voiding dysfunction.
Collapse
Affiliation(s)
- Anne E Turco
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison,Madison, WI 53705, USA
| | - Steven R Oakes
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kimberly P Keil Stietz
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cheryl L Dunham
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Diya B Joseph
- Department of Urology, University of Texas Southwestern, Dallas, TX 75390, USA
| | | | - Nicholas M Girardi
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Andrew J Schneider
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joseph Gawdzik
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Celeste M Sheftel
- Cellular and Molecular Pharmacology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Peiqing Wang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zunyi Wang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dale E Bjorling
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - William A Ricke
- Department of Urology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Weiping Tang
- Department of Urology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Laura L Hernandez
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Janet R Keast
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Adrian D Bonev
- Department of Pharmacology, University of Vermont, Burlington, VT 05405, USA
| | - Matthew D Grimes
- Department of Urology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Douglas W Strand
- Department of Urology, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Nathan R Tykocki
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 58823, USA
| | - Robyn L Tanguay
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Richard E Peterson
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison,Madison, WI 53705, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Chad M Vezina
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison,Madison, WI 53705, USA.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
10
|
Wegner KA, Ruetten H, Girardi NM, O’Driscoll CA, Sandhu JK, Turco AE, Abler LL, Wang P, Wang Z, Bjorling DE, Malinowski R, Peterson RE, Strand DW, Marker PC, Vezina CM. Genetic background but not prostatic epithelial beta-catenin influences susceptibility of male mice to testosterone and estradiol-induced urinary dysfunction. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2021; 9:121-131. [PMID: 33816700 PMCID: PMC8012832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
Urinary voiding dysfunction in aging men can cause bothersome symptoms and irreparable tissue damage. Underlying mechanisms are not fully known. We previously demonstrated that subcutaneous, slow-release testosterone and estradiol implants (T+E2) drive a pattern of urinary voiding dysfunction in male mice that resembles that of aging men. The initial goal of this study was to test the hypothesis that prostatic epithelial beta-catenin (Ctnnb1) is required for T+E2-mediated voiding dysfunction. Targeted Ctnnb1 deletion did not significantly change voiding function in control or T+E2 treated mice but led to the surprising discovery that the C57BL/6J × FVB/NJ × 129S1 mixed genetic background onto which Ctnnb1 loss of function alleles were maintained is profoundly susceptible to voiding dysfunction. The mixed background mice develop a more rapid T+E2-mediated increase in spontaneous urine spotting, are more impaired in ability to initiate bladder contraction, and develop larger and heavier bladders than T+E2 treated C57BL/6J pure bred mice. To better understand mechanisms, we separately evaluated contributions of T and E2 and found that E2 mediates voiding dysfunction. Our findings that genetic factors serve as modifiers of responsiveness to T and E2 demonstrate the need to control for genetic background in studies of male voiding dysfunction. We also show that genetic factors could control severity of voiding dysfunction. We demonstrate the importance of E2 as a key mediator of voiding impairment, and show that the concentration of E2 in subcutaneous implants determines the severity of voiding dysfunction in mice, demonstrating that the mouse model is tunable, a factor which is important for future pharmacological intervention studies.
Collapse
Affiliation(s)
- Kyle A Wegner
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Hannah Ruetten
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Nicholas M Girardi
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Chelsea A O’Driscoll
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Jaskiran K Sandhu
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Anne E Turco
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Lisa L Abler
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Peiqing Wang
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Zunyi Wang
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Dale E Bjorling
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Rita Malinowski
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
| | | | - Douglas W Strand
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- Department of Urology, University of Texas Southwestern Medical CenterDallas, Texas, USA
| | - Paul C Marker
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
| | - Chad M Vezina
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| |
Collapse
|
11
|
Keil Stietz KP, Kennedy CL, Sethi S, Valenzuela A, Nunez A, Wang K, Wang Z, Wang P, Spiegelhoff A, Puschner B, Bjorling DE, Lein PJ. In utero and lactational PCB exposure drives anatomic changes in the juvenile mouse bladder. Curr Res Toxicol 2021; 2:1-18. [PMID: 34337439 PMCID: PMC8317607 DOI: 10.1016/j.crtox.2021.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Bladder dysfunction, including incontinence, difficulty emptying the bladder, or urgency to urinate is a pervasive health and quality of life concern. However, risk factors for developing these symptoms are not completely understood, and the influence of exposure to environmental chemicals, especially during development, on the formation and function of the bladder is understudied. Environmental contaminants such as polychlorinated biphenyls (PCBs) are known to pose a risk to the developing brain; however, their influence on the development of peripheral target organs, such as bladder, are unknown. To address this data gap, C57Bl/6J mouse dams were exposed to an environmentally-relevant PCB mixture at 0, 0.1, 1 or 6 mg/kg daily beginning two weeks prior to mating and continuing through gestation and lactation. Bladders were collected from offspring at postnatal days (P) 28-31. PCB concentrations were detected in bladders in a dose-dependent manner. PCB effects on the bladder were sex- and dose-dependent. Overall, PCB effects were observed in male, but not female, bladders. PCBs increased bladder volume and suburothelial βIII-tubulin-positive nerve density compared to vehicle control. A subset of these nerves were sensory peptidergic axons indicated by increased calcitonin gene-related protein (CGRP) positive nerve fibers in mice exposed to the highest PCB dose compared to the lowest PCB dose. PCB-induced increased nerve density was also positively correlated with the number of mast cells in the bladder, suggesting inflammation may be involved. There were no detectable changes in epithelial composition or apoptosis as indicated by expression of cleaved caspase 3, suggesting PCBs do not cause overt toxicity. Bladder volume changes were not accompanied by changes in bladder mass or epithelial thickness, indicating that obstruction was not likely involved. Together, these results are the first to suggest that following developmental exposure, PCBs can distribute to the bladder and alter neuroanatomic development and bladder volume in male mice.
Collapse
Affiliation(s)
- Kimberly P. Keil Stietz
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, Davis, CA, USA,Department of Comparative Biosciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, USA,Corresponding author at: Department of Comparative Biosciences University of Wisconsin-Madison School of Veterinary Medicine, 2015 Linden Drive, Madison, WI 53706, USA.
| | - Conner L. Kennedy
- Department of Comparative Biosciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, USA
| | - Sunjay Sethi
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, Davis, CA, USA
| | - Anthony Valenzuela
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, Davis, CA, USA
| | - Alexandra Nunez
- Department of Comparative Biosciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, USA
| | - Kathy Wang
- Department of Comparative Biosciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, USA
| | - Zunyi Wang
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, USA
| | - Peiqing Wang
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, USA
| | - Audrey Spiegelhoff
- Department of Comparative Biosciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, USA
| | - Birgit Puschner
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, Davis, CA, USA
| | - Dale E. Bjorling
- Department of Surgical Sciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, Davis, CA, USA
| |
Collapse
|
12
|
Polychlorinated Biphenyls (PCBs): Risk Factors for Autism Spectrum Disorder? TOXICS 2020; 8:toxics8030070. [PMID: 32957475 PMCID: PMC7560399 DOI: 10.3390/toxics8030070] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Autism spectrum disorder (ASD) includes a group of multifactorial neurodevelopmental disorders defined clinically by core deficits in social reciprocity and communication, restrictive interests and repetitive behaviors. ASD affects one in 54 children in the United States, one in 89 children in Europe, and one in 277 children in Asia, with an estimated worldwide prevalence of 1-2%. While there is increasing consensus that ASD results from complex gene x environment interactions, the identity of specific environmental risk factors and the mechanisms by which environmental and genetic factors interact to determine individual risk remain critical gaps in our understanding of ASD etiology. Polychlorinated biphenyls (PCBs) are ubiquitous environmental contaminants that have been linked to altered neurodevelopment in humans. Preclinical studies demonstrate that PCBs modulate signaling pathways implicated in ASD and phenocopy the effects of ASD risk genes on critical morphometric determinants of neuronal connectivity, such as dendritic arborization. Here, we review human and experimental evidence identifying PCBs as potential risk factors for ASD and discuss the potential for PCBs to influence not only core symptoms of ASD, but also comorbidities commonly associated with ASD, via effects on the central and peripheral nervous systems, and/or peripheral target tissues, using bladder dysfunction as an example. We also discuss critical data gaps in the literature implicating PCBs as ASD risk factors. Unlike genetic factors, which are currently irreversible, environmental factors are modifiable risks. Therefore, data confirming PCBs as risk factors for ASD may suggest rational approaches for the primary prevention of ASD in genetically susceptible individuals.
Collapse
|
13
|
Taylor JA, Jones MB, Besch-Williford CL, Berendzen AF, Ricke WA, vom Saal FS. Interactive Effects of Perinatal BPA or DES and Adult Testosterone and Estradiol Exposure on Adult Urethral Obstruction and Bladder, Kidney, and Prostate Pathology in Male Mice. Int J Mol Sci 2020; 21:ijms21113902. [PMID: 32486162 PMCID: PMC7313472 DOI: 10.3390/ijms21113902] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
Obstructive voiding disorder (OVD) occurs during aging in men and is often, but not always, associated with increased prostate size, due to benign prostatic hyperplasia (BPH), prostatitis, or prostate cancer. Estrogens are known to impact the development of both OVD and prostate diseases, either during early urogenital tract development in fetal–neonatal life or later in adulthood. To examine the potential interaction between developmental and adult estrogen exposure on the adult urogenital tract, male CD-1 mice were perinatally exposed to bisphenol A (BPA), diethylstilbestrol (DES) as a positive control, or vehicle negative control, and in adulthood were treated for 4 months with Silastic capsules containing testosterone and estradiol (T+E2) or empty capsules. Animals exposed to BPA or DES during perinatal development were more likely than negative controls to have urine flow/kidney problems and enlarged bladders, as well as enlarged prostates. OVD in adult T+E2-treated perinatal BPA and DES animals was associated with dorsal prostate hyperplasia and prostatitis. The results demonstrate a relationship between elevated exogenous estrogen levels during urogenital system development and elevated estradiol in adulthood and OVD in male mice. These findings support the two-hit hypothesis for the development of OVD and prostate diseases.
Collapse
Affiliation(s)
- Julia A. Taylor
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA; (J.A.T.); (M.B.J.)
| | - Maren Bell Jones
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA; (J.A.T.); (M.B.J.)
| | | | - Ashley F. Berendzen
- Biomolecular Imaging Center, Harry S Truman VA Hospital and University of Missouri, Columbia, MO 65211, USA;
| | - William A. Ricke
- George M. O’Brien Center of Research Excellence, Department of Urology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA;
| | - Frederick S. vom Saal
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA; (J.A.T.); (M.B.J.)
- Correspondence: ; Tel.: +1-(573)-356-9621
| |
Collapse
|
14
|
Turco AE, Thomas S, Crawford LK, Tang W, Peterson RE, Li L, Ricke WA, Vezina CM. In utero and lactational 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD) exposure exacerbates urinary dysfunction in hormone-treated C57BL/6J mice through a non-malignant mechanism involving proteomic changes in the prostate that differ from those elicited by testosterone and estradiol. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2020; 8:59-72. [PMID: 32211455 PMCID: PMC7076297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 06/10/2023]
Abstract
A recent study directed new focus on the fetal and neonatal environment as a risk factor for urinary dysfunction in aging males. Male mice were exposed in utero and via lactation (IUL) to the persistent environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and then administered slow-release, subcutaneous implants of testosterone and estradiol (T+E2) as adults to mimic the hormonal environment of aging men. IUL TCDD exposure worsened T+E2-induced voiding dysfunction. Mice in the previous study were genetically prone to prostatic neoplasia and it was therefore unclear whether TCDD exacerbates voiding dysfunction through a malignant or non-malignant mechanism. We demonstrate here that IUL TCDD exposure acts via a non-malignant mechanism to exacerbate T+E2-mediated male mouse voiding dysfunction characterized by a progressive increase in spontaneous void spotting. We deployed a proteomic approach to narrow the possible mechanisms. We specifically tested whether IUL TCDD exacerbates urinary dysfunction by acting through the same prostatic signaling pathways as T+E2. The prostatic protein signature of TCDD/T+E2-exposed mice differed from that of mice exposed to T+E2 alone, indicating that the mechanism of action of TCDD differs from that of T+E2. We identified 3641 prostatic proteins in total and determined that IUL TCDD exposure significantly changed the abundance of 102 proteins linked to diverse molecular and physiological processes. We shed new light on the mechanism of IUL TCDD-mediated voiding dysfunction by demonstrating that the mechanism is independent of tumorigenesis and involves molecular pathways distinct from those affected by T+E2.
Collapse
Affiliation(s)
- Anne E Turco
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, WI, USA
- George M. O’Brien Center of Research Excellence, University of Wisconsin-MadisonMadison, WI, USA
| | - Samuel Thomas
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, WI, USA
- George M. O’Brien Center of Research Excellence, University of Wisconsin-MadisonMadison, WI, USA
- Department of Urology, University of Wisconsin-MadisonMadison, WI, USA
| | - LaTasha K Crawford
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI, USA
| | - Weiping Tang
- School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
| | - Richard E Peterson
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, WI, USA
- School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
| | - Lingjun Li
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, WI, USA
- School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
- Department of Chemistry, University of Wisconsin-MadisonMadison, WI, USA
| | - William A Ricke
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, WI, USA
- School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
- George M. O’Brien Center of Research Excellence, University of Wisconsin-MadisonMadison, WI, USA
- Department of Urology, University of Wisconsin-MadisonMadison, WI, USA
| | - Chad M Vezina
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, WI, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI, USA
- George M. O’Brien Center of Research Excellence, University of Wisconsin-MadisonMadison, WI, USA
- Department of Urology, University of Wisconsin-MadisonMadison, WI, USA
| |
Collapse
|
15
|
Mai X, Dong Y, Xiang L, Er Z. Maternal exposure to 2,3,7,8-tetrachlorodibenzo -p-dioxin suppresses male reproductive functions in their adulthood. Hum Exp Toxicol 2020; 39:890-905. [PMID: 32031418 DOI: 10.1177/0960327120903489] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a ubiquitous environmental contaminant in the environment. The developmental period is more sensitive to TCDD and there is a possibility that maternal exposure to TCDD may affect in adulthood. Adult female Wistar rats were exposed to 0.5, 1.0, and 2.0 µg/kg TCDD during the critical stage of organogenesis, that is, on GD15. The results revealed a significant decrease in indices of reproductive organ weight in adult male rats exposed to prenatal TCDD, and dose-dependent reduction in epididymal sperm reserves, percent motile, and viable sperm with an increase in percent morphological abnormal sperm. Polymerase chain reaction analysis revealed downregulated expression levels of steroidogenic markers such as steroidogenic acute regulatory, cholesterol side-chain cleavage, and 3β- and 17β-hydroxysteroid dehydrogenase (HSDs) in experimental rats. Immunofluorescence sections portrayed reduced distribution of 3β- and 17β-HSD proteins in testes of experimental rats. Furthermore, spermatogenic markers (acid phosphatase, alkaline phosphatase, lactate dehydrogenase, and sorbitol dehydrogenase) were significantly altered in the testes. Serum levels of testosterone, follicle stimulating hormones, and luteinizing hormone were significantly decreased. Testicular levels of hydrogen peroxide and lipid peroxidation were significantly elevated with a decline in superoxide dismutase, catalase, glutathione peroxidase activities, and total thiol levels. Moreover, histological and morphometric examination of testicular cross-sections depicted degenerative changes. Male fertility assessment in adult rats revealed a significant decrease in mating index, fertility index, and mean number of pre- and postimplantations with an increase in pre- and postimplantation losses in rats cohabited with in utero TCDD-exposed adult males. In conclusion, the findings of this study provided clear evidence that maternal exposure to TCDD during the critical stage of development results in suppressed reproductive health in adulthood.
Collapse
Affiliation(s)
- X Mai
- Department of Reproductive medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Y Dong
- Department of Reproductive medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - L Xiang
- Department of Reproductive medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Z Er
- Department of Reproductive medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
16
|
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
|
17
|
Liu TT, Rodgers AC, Nicholson TM, Macoska JA, Marker PC, Vezina CM, Bjorling DE, Roldan-Alzate A, Hernando D, Lloyd GL, Hacker TA, Ricke WA. Ultrasonography of the Adult Male Urinary Tract for Urinary Functional Testing. J Vis Exp 2019:10.3791/59802. [PMID: 31475976 PMCID: PMC7328372 DOI: 10.3791/59802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The incidence of clinical benign prostatic hyperplasia (BPH) and lower urinary tract symptoms (LUTS) is increasing due to the aging population, resulting in a significant economic and quality of life burden. Transgenic and other mouse models have been developed to recreate various aspects of this multifactorial disease; however, methods to accurately quantitate urinary dysfunction and the effectiveness of new therapeutic options are lacking. Here, we describe a method that can be used to measure bladder volume and detrusor wall thickness, urinary velocity, void volume and void duration, and urethral diameter. This would allow for the evaluation of disease progression and treatment efficacy over time. Mice were anesthetized with isoflurane, and the bladder was visualized by ultrasound. For non-contrast imaging, a 3D image was taken of the bladder to calculate volume and evaluate shape; the bladder wall thickness was measured from this image. For contrast-enhanced imaging, a catheter was placed through the dome of the bladder using a 27-gauge needle connected to a syringe by PE50 tubing. A bolus of 0.5 mL of contrast was infused into the bladder until a urination event occurred. Urethral diameter was determined at the point of the Doppler velocity sample window during the first voiding event. Velocity was measured for each subsequent event yielding a flow rate. In conclusion, high frequency ultrasound proved to be an effective method for assessing bladder and urethral measurements during urinary function in mice. This technique may be useful in the assessment of novel therapies for BPH/LUTS in an experimental setting.
Collapse
Affiliation(s)
- Teresa T Liu
- Department of Urology, University of Wisconsin-Madison; K12 Kure, University of Wisconsin-Madison
| | - Allison C Rodgers
- Cardiovascular Research Center, Department of Medicine, University of Wisconsin-Madison
| | | | - Jill A Macoska
- University of Massachusetts Boston; U54 George M. O'Brien Center, University of Wisconsin-Madison
| | - Paul C Marker
- U54 George M. O'Brien Center, University of Wisconsin-Madison; College of Pharmacy, University of Wisconsin-Madison
| | - Chad M Vezina
- U54 George M. O'Brien Center, University of Wisconsin-Madison; School of Veterinary Medicine, University of Wisconsin-Madison
| | - Dale E Bjorling
- U54 George M. O'Brien Center, University of Wisconsin-Madison; School of Veterinary Medicine, University of Wisconsin-Madison
| | - Alejandro Roldan-Alzate
- K12 Kure, University of Wisconsin-Madison; Department of Mechanical Engineering, University of Wisconsin-Madison; Department of Radiology, University of Wisconsin-Madison
| | - Diego Hernando
- Department of Radiology, University of Wisconsin-Madison; Department of Medical Physics, University of Wisconsin-Madison
| | | | - Timothy A Hacker
- Cardiovascular Research Center, Department of Medicine, University of Wisconsin-Madison
| | - William A Ricke
- Department of Urology, University of Wisconsin-Madison; U54 George M. O'Brien Center, University of Wisconsin-Madison;
| |
Collapse
|
18
|
Ruetten H, Wegner KA, Zhang HL, Wang P, Sandhu J, Sandhu S, Mueller B, Wang Z, Macoska J, Peterson RE, Bjorling DE, Ricke WA, Marker PC, Vezina CM. Impact of sex, androgens, and prostate size on C57BL/6J mouse urinary physiology: functional assessment. Am J Physiol Renal Physiol 2019; 317:F996-F1009. [PMID: 31390231 DOI: 10.1152/ajprenal.00270.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Laboratory mice are used to identify causes of urinary dysfunction including prostate-related mechanisms of lower urinary tract symptoms. Effective use of mice for this purpose requires a clear understanding of molecular, cellular, anatomic, and endocrine contributions to voiding function. Whether the prostate influences baseline voiding function has not been specifically evaluated, in part because most methods that alter prostate mass also change circulating testosterone concentrations. We performed void spot assay and cystometry to establish a multiparameter "baseline" of voiding function in intact male and female 9-wk-old (adult) C57BL/6J mice. We then compared voiding function in intact male mice to that of castrated male mice, male (and female) mice treated with the steroid 5α-reductase inhibitor finasteride, or male mice harboring alleles (Pbsn4cre/+; R26RDta/+) that significantly reduce prostate lobe mass by depleting prostatic luminal epithelial cells. We evaluated aging-related changes in male urinary voiding. We also treated intact male, castrate male, and female mice with exogenous testosterone to determine the influence of androgen on voiding function. The three methods used to reduce prostate mass (castration, finasteride, and Pbsn4cre/+; R26RDta/+) changed voiding function from baseline but in a nonuniform manner. Castration feminized some aspects of male urinary physiology (making them more like intact female mice) while exogenous testosterone masculinized some aspects of female urinary physiology (making them more like intact male mice). Our results provide evidence that circulating testosterone is responsible in part for baseline sex differences in C57BL/6J mouse voiding function while prostate lobe mass in young, healthy adult mice has a lesser influence.
Collapse
Affiliation(s)
- Hannah Ruetten
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts
| | - Kyle A Wegner
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts.,Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Helen L Zhang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts
| | - Peiqing Wang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts.,Center for Personalized Cancer Therapy, The University of Massachusetts Boston, Boston, Massachusetts
| | - Jaskiran Sandhu
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts
| | - Simran Sandhu
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts
| | - Brett Mueller
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts
| | - Zunyi Wang
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts.,Center for Personalized Cancer Therapy, The University of Massachusetts Boston, Boston, Massachusetts
| | - Jill Macoska
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts.,Center for Personalized Cancer Therapy, The University of Massachusetts Boston, Boston, Massachusetts
| | - Richard E Peterson
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Dale E Bjorling
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts.,Department of Surgical Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - William A Ricke
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts.,Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Urology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Paul C Marker
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts.,Center for Personalized Cancer Therapy, The University of Massachusetts Boston, Boston, Massachusetts
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin, and Boston, Massachusetts.,Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
19
|
Yoshioka W, Tohyama C. Mechanisms of Developmental Toxicity of Dioxins and Related Compounds. Int J Mol Sci 2019; 20:E617. [PMID: 30708991 PMCID: PMC6387164 DOI: 10.3390/ijms20030617] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Dioxins and related compounds induce morphological abnormalities in developing animals in an aryl hydrocarbon receptor (AhR)-dependent manner. Here we review the studies in which 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is used as a prototypical compound to elucidate the pathogenesis of morphological abnormalities. TCDD-induced cleft palate in fetal mice involves a delay in palatogenesis and dissociation of fused palate shelves. TCDD-induced hydronephrosis, once considered to be caused by the anatomical obstruction of the ureter, is now separated into TCDD-induced obstructive and non-obstructive hydronephrosis, which develops during fetal and neonatal periods, respectively. In the latter, a prostaglandin E₂ synthesis pathway and urine concentration system are involved. TCDD-induced abnormal development of prostate involves agenesis of the ventral lobe. A suggested mechanism is that AhR activation in the urogenital sinus mesenchyme by TCDD modulates the wingless-type MMTV integration site family (WNT)/β-catenin signaling cascade to interfere with budding from urogenital sinus epithelium. TCDD exposure to zebrafish embryos induces loss of epicardium progenitor cells and heart malformation. AHR2-dependent downregulation of Sox9b expression in cardiomyocytes is a suggested underlying mechanism. TCDD-induced craniofacial malformation in zebrafish is considered to result from the AHR2-dependent reduction in SRY-box 9b (SOX9b), probably partly via the noncoding RNA slincR, resulting in the underdevelopment of chondrocytes and cartilage.
Collapse
Affiliation(s)
- Wataru Yoshioka
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
20
|
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
|
21
|
Cunha GR, Vezina CM, Isaacson D, Ricke WA, Timms BG, Cao M, Franco O, Baskin LS. Development of the human prostate. Differentiation 2018; 103:24-45. [PMID: 30224091 PMCID: PMC6234090 DOI: 10.1016/j.diff.2018.08.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/14/2022]
Abstract
This paper provides a detailed compilation of human prostatic development that includes human fetal prostatic gross anatomy, histology, and ontogeny of selected epithelial and mesenchymal differentiation markers and signaling molecules throughout the stages of human prostatic development: (a) pre-bud urogenital sinus (UGS), (b) emergence of solid prostatic epithelial buds from urogenital sinus epithelium (UGE), (c) bud elongation and branching, (d) canalization of the solid epithelial cords, (e) differentiation of luminal and basal epithelial cells, and (f) secretory cytodifferentiation. Additionally, we describe the use of xenografts to assess the actions of androgens and estrogens on human fetal prostatic development. In this regard, we report a new model of de novo DHT-induction of prostatic development from xenografts of human fetal female urethras, which emphasizes the utility of the xenograft approach for investigation of initiation of human prostatic development. These studies raise the possibility of molecular mechanistic studies on human prostatic development through the use of tissue recombinants composed of mutant mouse UGM combined with human fetal prostatic epithelium. Our compilation of human prostatic developmental processes is likely to advance our understanding of the pathogenesis of benign prostatic hyperplasia and prostate cancer as the neoformation of ductal-acinar architecture during normal development is shared during the pathogenesis of benign prostatic hyperplasia and prostate cancer.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States.
| | - Chad M Vezina
- School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, United States
| | - Dylan Isaacson
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - William A Ricke
- Department of Urology, University of Wisconsin, Madison, WI 53705, United States
| | - Barry G Timms
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, United States
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - Omar Franco
- Department of Surgery, North Shore University Health System, 1001 University Place, Evanston, IL 60201, United States
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| |
Collapse
|
22
|
Hill WG, Zeidel ML, Bjorling DE, Vezina CM. Void spot assay: recommendations on the use of a simple micturition assay for mice. Am J Physiol Renal Physiol 2018; 315:F1422-F1429. [PMID: 30156116 DOI: 10.1152/ajprenal.00350.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Investigators have for decades used mouse voiding patterns as end points for studying behavioral biology. It is only recently that mouse voiding patterns were adopted for study of lower urinary tract physiology. The spontaneous void spot assay (VSA), a popular micturition assessment tool, involves placing a mouse in an enclosure lined by filter paper and quantifying the resulting urine spot pattern. The VSA has advantages of being inexpensive and noninvasive, but some investigators challenge its ability to distinguish lower urinary tract function from behavioral voiding. A consensus group of investigators who regularly use the VSA was established by the National Institutes of Health in 2015 to address the strengths and weaknesses of the assay, determine whether it can be standardized across laboratories, and determine whether it can be used as a surrogate for evaluating urinary function. Here we leverage experience from the consensus group to review the history of the VSA and its uses, summarize experiments to optimize assay design for urinary physiology assessment, and make best practice recommendations for performing the assay and analyzing its results.
Collapse
Affiliation(s)
- Warren G Hill
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| | - Mark L Zeidel
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| | - Dale E Bjorling
- Department of Surgical Sciences, University of Wisconsin-Madison , Madison, Wisconsin.,University of Wisconsin-Madison/University of Massachusetts-Boston, George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin and Boston, Massachusetts
| | - Chad M Vezina
- University of Wisconsin-Madison/University of Massachusetts-Boston, George M. O'Brien Center for Benign Urologic Research, Madison, Wisconsin and Boston, Massachusetts.,Department of Comparative Biosciences, University of Wisconsin-Madison , Madison, Wisconsin
| |
Collapse
|
23
|
Xia B, Wang Y, Wang X, Wu J, Song Q, Sun Z, Zhang Y. In utero and lactational exposure of DEHP increases the susceptibility of prostate carcinogenesis in male offspring through PSCA hypomethylation. Toxicol Lett 2018; 292:78-84. [PMID: 29689378 DOI: 10.1016/j.toxlet.2018.04.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/27/2018] [Accepted: 04/20/2018] [Indexed: 12/17/2022]
Abstract
As an ubiquitous environmental endocrine disruptor, di(2-ethylhexyl) phthalate (DEHP) has been shown to interfere with the development of reproductive organs and induce pathological changes in prostate. Our previous finding showed that in utero and lactational (IUL) DEHP exposure could disrupt the balance of testosterone and estrogen and increase the susceptibility of prostate carcinogenesis. The purpose of this study is to investigate whether the early-life specific epigenetic modifications could mediate the effect of DEHP exposure on prostate carcinogenesis in rodents, for epigenetic modifications play important roles in regulating prostate carcinogenesis. The pregnant rats were treated with corn oil (negative control) or DEHP at 0.01, 0.1 and 1 mg/kg BW/day from GD7 to PND21. On PND21, the expression and DNA methylation change of six prostate carcinogenesis-related genes (ESR2/GSTP1/NKX3.1/PSCA/PTGS2/Rassf1a) were assessed through SYBR-Green real-time PCR combined with pyrosequencing assay in F1 male offspring. On PND196, the relationship b(STP1, PSCA and PTGS2 in a dose-dependent manner, which were positively correlated with PIN scores, Gleason scores, serum PSA concentrations and negatively correlated with prostate/body weight ratio on PND196. Meanwhile, 1 mg/kg BW/day DEHP markedly reduced DNA methylation level of PSCA in all studied CpG sites. Significant inverse correlations between methylation levels of the promoter CpG site and PSCA mRNA expression were observed. These results indicated that transcriptional changes of GSTP1, PSCA and PTGS2 induced by DEHP exposure might be contribute to the increasing susceptibility of prostate carcinogenesis in late life. Moreover, hypomethylation of PSCA could mediate the effect of DEHP on prostate carcinogenesis in rats.
Collapse
Affiliation(s)
- Bin Xia
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yong Wang
- WHO Collaborating Center for Research in Human Reproduction, Shanghai Institute of Planned Parenthood Research, Shanghai 200030, China
| | - Xiu Wang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jianhui Wu
- WHO Collaborating Center for Research in Human Reproduction, Shanghai Institute of Planned Parenthood Research, Shanghai 200030, China
| | - Qi Song
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
| | - Zuyue Sun
- WHO Collaborating Center for Research in Human Reproduction, Shanghai Institute of Planned Parenthood Research, Shanghai 200030, China.
| | - Yunhui Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
24
|
Wegner KA, Keikhosravi A, Eliceiri KW, Vezina CM. Fluorescence of Picrosirius Red Multiplexed With Immunohistochemistry for the Quantitative Assessment of Collagen in Tissue Sections. J Histochem Cytochem 2017; 65:479-490. [PMID: 28692327 DOI: 10.1369/0022155417718541] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The low cost and simplicity of picrosirius red (PSR) staining have driven its popularity for collagen detection in tissue sections. We extended the versatility of this method by using fluorescent imaging to detect the PSR signal and applying automated quantification tools. We also developed the first PSR protocol that is fully compatible with multiplex immunostaining, making it possible to test whether collagen structure differs across immunohistochemically labeled regions of the tissue landscape. We compared our imaging method with two gold standards in collagen imaging, linear polarized light microscopy and second harmonic generation imaging, and found that it is at least as sensitive and robust to changes in sample orientation. As proof of principle, we used a genetic approach to overexpress beta catenin in a patchy subset of mouse prostate epithelial cells distinguished only by immunolabeling. We showed that collagen fiber length is significantly greater near beta catenin overexpressing cells than near control cells. Our fluorescent PSR imaging method is sensitive, reproducible, and offers a new way to guide region of interest selection for quantifying collagen in tissue sections.
Collapse
Affiliation(s)
- Kyle A Wegner
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin (KAW).,George M. O'Brien Research Center of Excellence, University of Wisconsin-Madison, Madison, Wisconsin (KAW, CMV, KWE)
| | - Adib Keikhosravi
- Department of Biomedical Engineering (AK, KWE), Madison, Wisconsin.,Laboratory for Optical and Computational Instrumentation (AK, KWE), Madison, Wisconsin
| | - Kevin W Eliceiri
- Department of Biomedical Engineering (AK, KWE), Madison, Wisconsin.,Laboratory for Optical and Computational Instrumentation (AK, KWE), Madison, Wisconsin.,Morgridge Institute for Research, Madison, Wisconsin (KWE).,Comprehensive Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin (KWE).,George M. O'Brien Research Center of Excellence, University of Wisconsin-Madison, Madison, Wisconsin (KAW, CMV, KWE)
| | - Chad M Vezina
- Department of Comparative Biosciences (CMV), Madison, Wisconsin.,George M. O'Brien Research Center of Excellence, University of Wisconsin-Madison, Madison, Wisconsin (KAW, CMV, KWE)
| |
Collapse
|