Clinical significance of steroid and xenobiotic receptor and its targeted gene CYP3A4 in human prostate cancer,

2020 ◽  
Author(s):  
Lungwani Muungo

The steroid and xenobiotic receptor (SXR) regulates cytochromeP450 (CYP) enzymes, which are key inactivators of testosterone inthe liver and prostate. In the present study, we investigated SXRexpression in human prostate tissues. We determined SXR immunoreactivityusing an anti-SXR antibody in benign (n = 78) andcancerous (n = 106) tissues obtained by radical prostatectomy.Stained slides were evaluated for the proportion and stainingintensity of immunoreactive cells. Total immunoreactivity (IR)scores (range: 0–8) were calculated as the sum of the proportionand intensity scores. Associations between the clinicopathologicalfeatures of the patients, SXR status, and CYP3A4 immunoreactivitywere analyzed. Western blot analyses validated the specificity ofthe anti-SXR antibody in 293T cells transfected with pcDNA–FLAG–SXR. Positive (IR score: ‡ 2) nuclear SXR staining was observed in91% (71 ⁄ 78) of benign foci and 47% (50 ⁄ 106) of cancerous lesions.Immunoreactivity scores were significantly lower in the cancerouslesions than in the benign foci (P < 0.0001). Clinicopathologicalanalyses showed that cancer-specific survival in patients with highSXR IR scores (‡4) was significantly increased (P = 0.046). Combineddata of present and previous studies showed that high IRscores for both the SXR and CYP3A4 correlated with significantlybetter cancer-specific survival rates in multivariate regression analyses(hazard ratio: 2.15, 95% confidence interval: 1.25–3.55,P = 0.007). We showed differential SXR expression in human prostatetissues. The high expression of the SXR and CYP3A4 is astrong prognostic indicator of favorable outcomes in prostatecancer, and could be a therapeutic target. (Cancer Sci 2012; 103:176–180)Endocrine therapies for advanced or recurrent prostate cancer(PC) are intended to decrease levels of circulatingandrogens and ⁄ or reduce androgen receptor (AR) activity.(1)Unfortunately, PC frequently develops into castration-resistantprostate cancer (CRPC). In CRPC, AR activity is often preservedby various mechanisms, including changes in intratumorligand concentration, AR gene amplification, AR mutations,changes in regulatory molecules, and ligand-independent ARactivation through kinase cross-talk.(2) Additionally, CRPCresults in high androstenediol levels in PC tissues, even afterandrogen-deprivation therapy.(3) Androstenediol is a morepotent activator of the mutant AR than dihydrotestosterone(DHT) in LNCaP cells, and induces greater cell proliferation.(3)Moreover, cells of a hormone-refractory MDA–PCa-2b subline,which is a metastasis-derived subline generated in vitro frombone, are stimulated by low levels of testosterone.(4) We alsoproposed a possible correlation between PC and intraprostaticandrogen metabolism.(5) In our previous study, decreased cytochromeP450 (CYP)2B6 expression contributed to the progressionof human PC, and transfection of the CYP2B6 geneinhibited proliferation of LNCaP cells.(5) Dihydrotestosterone isinactivated by the enzyme CYP3A4, which is a member of theCYP450 family and is responsible for the hydroxylation of testosteronein the liver and prostate.(6–11) In our previous study,decreased CYP3A4 expression was associated with a higherGleason score (GS) and significantly decreased cancer-specificsurvival in human PC.(12) Therefore, decreased CYP3A4 activitymight contribute to the increased bioavailability of intraprostaticDHT and the subsequent development of PC.Cytochrome P450 3A4 expression is regulated by the steroidand xenobiotic receptor (SXR), also known as the human pregnaneX receptor, which consists of members of the NR1I nuclearreceptor subfamily of ligand-activated transcription factors.(13,14)The SXR is expressed primarily in the liver, intestine, kidney,lung, stomach, peripheral blood monocytes, blood–brain barrier,uterus, ovary, placenta, breast, osteoclasts, heart, adrenal gland,bone marrow, and specific regions of the brain.(13) This wideexpression pattern indicates that the SXR is part of a protectivemechanism for critical cells that are sensitive to aberrant levels ofexogenous or endogenous compounds, such as phenobarbital,dexamethasone (DXM), prednisolone (PSL), androstane, andestrogens.(13) Recent studies have also revealed SXR-activated,signal-regulated proliferation in breast and colon cancer.(15,16)Therefore, we hypothesized that the SXR, which is upstream ofthe CYP3A4 gene, might be associated with PC progression. Inthe present study, we investigated the SXR expression in humanprostate tissues using an anti-SXR antibody.

2011 ◽  
Vol 103 (2) ◽  
pp. 176-180 ◽  
Author(s):  
Tetsuya Fujimura ◽  
Satoru Takahashi ◽  
Tomohiko Urano ◽  
Toshiya Tanaka ◽  
Weiguang Zhang ◽  
...  

2008 ◽  
Vol 99 (7) ◽  
pp. 1362-1369 ◽  
Author(s):  
Weidong Hu ◽  
Xinming Zhen ◽  
Bin Xiong ◽  
Bicheng Wang ◽  
Weibing Zhang ◽  
...  

Nutrients ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 1000
Author(s):  
Lu Yu ◽  
Robert W. Li ◽  
Haiqiu Huang ◽  
Quynhchi Pham ◽  
Liangli Yu ◽  
...  

LNCaP athymic xenograft model has been widely used to allow researchers to examine the effects and mechanisms of experimental treatments such as diet and diet-derived cancer preventive and therapeutic compounds on prostate cancer. However, the biological characteristics of human LNCaP cells before/after implanting in athymic mouse and its relevance to clinical human prostate outcomes remain unclear and may dictate interpretation of biological efficacies/mechanisms of diet/diet-derived experimental treatments. In this study, transcriptome profiles and pathways of human prostate LNCaP cells before (in vitro) and after (in vivo) implanting into xenograft mouse were compared using RNA-sequencing technology (RNA-seq) followed by bioinformatic analysis. A shift from androgen-responsive to androgen nonresponsive status was observed when comparing LNCaP xenograft tumor to culture cells. Androgen receptor and aryl-hydrocarbon pathway were found to be inhibited and interleukin-1 (IL-1) mediated pathways contributed to these changes. Coupled with in vitro experiments modeling for androgen exposure, cell-matrix interaction, inflammation, and hypoxia, we identified specific mechanisms that may contribute to the observed changes in genes and pathways. Our results provide critical baseline transcriptomic information for a tumor xenograft model and the tumor environments that might be associated with regulating the progression of the xenograft tumor, which may influence interpretation of diet/diet-derived experimental treatments.


2020 ◽  
Author(s):  
Lungwani Muungo

The androgen receptor (AR) plays a critical role in the development and the progression of prostate cancer. Alterations in theexpression of AR coregulators lead to AR hypersensitivity, which is one of the mechanisms underlying the progression ofprostate cancer into a castrate-resistant state. Octamer transcription factor 1 (Oct1) is a ubiquitous member of the POUhomeodomainfamily that functions as a coregulator of AR. In our study, the contribution of Oct1 to prostate cancerdevelopment was examined. Immunocytochemistry analysis showed that Oct1 is expressed in the nuclei of LNCaP cells.siRNA-mediated silencing of Oct1 expression inhibited LNCaP cell proliferation. Immunohistochemical analysis of Oct1expression in tumor specimens obtained from 102 patients with prostate cancer showed a positive correlation of Oct1immunoreactivity with a high Gleason score and AR immunoreactivity (p 5 0.0042 and p < 0.0001, respectively). Moreover,patients with high immunoreactivity of Oct1 showed a low cancer-specific survival rate, and those patients with highimmunoreactivities of both Oct1 and AR exhibited poorer cancer-specific prognosis. Multivariate hazard analysis revealed asignificant correlation between high Oct1 immunoreactivity and poor cancer-specific survival (p 5 0.012). These resultsdemonstrate that Oct1 can be a prognostic factor in prostate cancer as a coregulator of AR and may lead to the developmentof a new therapeutic intervention for prostate cancer.


Molecules ◽  
2021 ◽  
Vol 26 (11) ◽  
pp. 3268
Author(s):  
Katja S. Håheim ◽  
Emil Lindbäck ◽  
Kah Ni Tan ◽  
Marte Albrigtsen ◽  
Ida T. Urdal Helgeland ◽  
...  

A series of novel quinoline-based tetracyclic ring-systems were synthesized and evaluated in vitro for their antiplasmodial, antiproliferative and antimicrobial activities. The novel hydroiodide salts 10 and 21 showed the most promising antiplasmodial inhibition, with compound 10 displaying higher selectivity than the employed standards. The antiproliferative assay revealed novel pyridophenanthridine 4b to be significantly more active against human prostate cancer (IC50 = 24 nM) than Puromycin (IC50 = 270 nM) and Doxorubicin (IC50 = 830 nM), which are used for clinical treatment. Pyridocarbazoles 9 was also moderately effective against all the employed cancer cell lines and moreover showed excellent biofilm inhibition (9a: MBIC = 100 µM; 9b: MBIC = 100 µM).


2012 ◽  
Vol 22 (17) ◽  
pp. 5470-5474 ◽  
Author(s):  
Byung Jun Ryu ◽  
Seung-hwa Baek ◽  
Jiyeon Kim ◽  
Su Jung Bae ◽  
Sung-Youn Chang ◽  
...  

2016 ◽  
Vol 11 (2) ◽  
pp. 378
Author(s):  
Jin-Jun Sun ◽  
Shi-Feng Kan ◽  
Guan-Xing Sun

<p class="Abstract">We tried a new method of prostate cancer treatment by inducing<em> in vitro</em> differentiation which resulted in reduction of cancer cells growth. A protein kinase inhibitor, midostaurin's ability to trigger the human prostate cancer cell line, DU145 to segregate into nerve cells was studied. Midostaurin (100 nM) suppressed the growth of DU145 cells but without change in the number of dead cells. Midostaurin started to extend neurites on DU145 cells after 24 hours and differentiated into nerve cells by 72 hours. The microtubule was stabilized by tau protein and its mRNA expression showed time-dependent increase in midostaurin-treated DU145 cells. At the same time, the amount of acetylcholinesterase was also increased. The midostaurin-treated DU145 cells showed 40% less activity than control in the colony forming assay. The results suggests that midostaurin can induce differentiation of DU145 cells into nerve cells.</p><p> </p>


2021 ◽  
Author(s):  
Peace C. Asuzu ◽  
Alberta N.A. Aryee ◽  
Nicholas Trompeter ◽  
Yasmin Mann ◽  
Samuel A. Besong ◽  
...  

AbstractPhenolic compounds are products of secondary plant metabolism known for their biological activity including their antimicrobial, antioxidant, analgesic, stimulant, anti- carcinogenic, and aphrodisiac properties. The main objective of this study was to assess the potency/cytotoxic effects of Prunus africana extracts on prostate cancer cells in vitro. Using different concentrations of P. africana extracts, prostate cancer C4-2 cells, a hormonally insensitive subline of LNCaP cells, were treated in a proliferation assay. A concentration dependent inhibition of cell growth in cells treated with P. africana bark and root extracts was present from days 1 through 3 of incubation, with the methanol extract of the bark showing the strongest effect. Compared to other plant parts, leaf extracts were significantly less cytotoxic at the same concentrations. As C4-2 cells are hormonally insensitive and designed to mimic advanced prostate cancer, crude extracts of P. africana are a possible treatment option, not only for hormone sensitive prostate cancer, but also advanced, hormonally insensitive prostate cancer.


Sign in / Sign up

Export Citation Format

Share Document