scholarly journals Exploring the Value of BRD9 as a Biomarker, Therapeutic Target and Co-Target in Prostate Cancer

Biomolecules ◽  
2021 ◽  
Vol 11 (12) ◽  
pp. 1794
Author(s):  
Nafisa Barma ◽  
Timothy C. Stone ◽  
Lina Maria Carmona Echeverria ◽  
Susan Heavey

Background and aims: Despite recent advances in advanced prostate cancer treatments, clinical biomarkers or treatments for men with such cancers are imperfect. Targeted therapies have shown promise, but there remain fewer actionable targets in prostate cancer than in other cancers. This work aims to characterise BRD9, currently understudied in prostate cancer, and investigate its co-expression with other genes to assess its potential as a biomarker and therapeutic target in human prostate cancer. Materials and methods: Omics data from a total of 2053 prostate cancer patients across 11 independent datasets were accessed via Cancertool and cBioPortal. mRNA M.expression and co-expression, mutations, amplifications, and deletions were assessed with respect to key clinical parameters including survival, Gleason grade, stage, progression, and treatment. Network and pathway analysis was carried out using Genemania, and heatmaps were constructed using Morpheus. Results: BRD9 is overexpressed in prostate cancer patients, especially those with metastatic disease. BRD9 expression did not differ in patients treated with second generation antiandrogens versus those who were not. BRD9 is co-expressed with many genes in the SWI/SNF and BET complexes, as well as those in common signalling pathways in prostate cancer. Summary and conclusions: BRD9 has potential as a diagnostic and prognostic biomarker in prostate cancer. BRD9 also shows promise as a therapeutic target, particularly in advanced prostate cancer, and as a co-target alongside other genes in the SWI/SNF and BET complexes, and those in common prostate cancer signalling pathways. These promising results highlight the need for wider experimental inhibition and co-targeted inhibition of BRD9 in vitro and in vivo, to build on the limited inhibition data available.

Author(s):  
Nafisa Barma ◽  
Timothy C Stone ◽  
Lina Maria Carmona Echeverria ◽  
Susan Heavey

Background and aims: Despite recent advances in advanced prostate cancer treatments, there are no clinically useful biomarkers or treatments for men with such cancers. Targeted therapies have shown promise, but there remain fewer actionable targets in prostate cancer than in other cancers. This work aims to characterize BRD9, currently understudied in prostate cancer, and investigate its co-expression with other genes to assess its potential as a biomarker and therapeutic target in human prostate cancer. Materials and methods: Omics data from a total of 2053 prostate cancer patients across 11 independent datasets were accessed via Cancertool and cBioPortal. mRNA expression and co-expression, mutations, amplifications, and deletions were assessed with respect to key clinical parameters including survival, Gleason grade, stage, progression and treatment. Network and pathway analysis was carried out using Genemania, and heatmaps were constructed using Morpheus. Results: BRD9 is overexpressed in prostate cancer patients, especially those with metastatic disease. BRD9 expression did not differ in patients treated with second generation antiandrogens versus those who were not. BRD9 is co-expressed with many genes in the SWI/SNF and BET complexes, as well as those in common signaling pathways in prostate cancer. Summary and conclusions: BRD9 has potential as a diagnostic and prognostic biomarker in prostate cancer. BRD9 also shows promise as a therapeutic target, particularly in advanced prostate cancer, and as a co-target alongside other genes in the SWI/SNF and BET complexes, and those in common prostate cancer signalling pathways. These promising results highlight the need for wider experimental inhibition and co-targeted inhibition of BRD9 in vitro and in vivo, to build on the limited inhibition data available.


Tumor Biology ◽  
2018 ◽  
Vol 40 (4) ◽  
pp. 101042831877177 ◽  
Author(s):  
Andrea Mancini ◽  
Alessandro Colapietro ◽  
Simona Pompili ◽  
Andrea Del Fattore ◽  
Simona Delle Monache ◽  
...  

Morbidity in advanced prostate cancer patients is largely associated with bone metastatic events. The development of novel therapeutic strategies is imperative in order to effectively treat this incurable stage of the malignancy. In this context, Akt signaling pathway represents a promising therapeutic target able to counteract biochemical recurrence and metastatic progression in prostate cancer. We explored the therapeutic potential of a novel dual PI3 K/mTOR inhibitor, X480, to inhibit tumor growth and bone colonization using different in vivo prostate cancer models including the subcutaneous injection of aggressive and bone metastatic (PC3) and non-bone metastatic (22rv1) cell lines and preclinical models known to generate bone lesions. We observed that X480 both inhibited the primary growth of subcutaneous tumors generated by PC3 and 22rv1 cells and reduced bone spreading of PCb2, a high osteotropic PC3 cell derivative. In metastatic bone, X480 inhibited significantly the growth and osteolytic activity of PC3 cells as observed by intratibial injection model. X480 also increased the bone disease-free survival compared to untreated animals. In vitro experiments demonstrated that X480 was effective in counteracting osteoclastogenesis whereas it stimulated osteoblast activity. Our report provides novel information on the potential activity of PI3 K/Akt inhibitors on the formation and progression of prostate cancer bone metastases and supports a biological rationale for the use of these inhibitors in castrate-resistant prostate cancer patients at high risk of developing clinically evident bone lesions.


2019 ◽  
Vol 8 (12) ◽  
pp. 2056 ◽  
Author(s):  
Juan M. Jiménez-Vacas ◽  
Enrique Gómez-Gómez ◽  
Antonio J. Montero-Hidalgo ◽  
Vicente Herrero-Aguayo ◽  
Fernando L-López ◽  
...  

Recent data suggested that plasma Ghrelin O-Acyl Transferase enzyme (GOAT) levels could represent a new diagnostic biomarker for prostate cancer (PCa). In this study, we aimed to explore the diagnostic and prognostic/aggressiveness capacity of GOAT in urine, as well as to interrogate its putative pathophysiological role in PCa. We analysed urine/plasma levels of GOAT in a cohort of 993 patients. In vitro (i.e., cell-proliferation) and in vivo (tumor-growth in a xenograft-model) approaches were performed in response to the modulation of GOAT expression/activity in PCa cells. Our results demonstrate that plasma and urine GOAT levels were significantly elevated in PCa patients compared to controls. Remarkably, GOAT significantly outperformed PSA in the diagnosis of PCa and significant PCa in patients with PSA levels ranging from 3 to 10 ng/mL (the so-called PSA grey-zone). Additionally, urine GOAT levels were associated to clinical (e.g., Gleason-score, PSA levels) and molecular (e.g., CDK2/CDK6/CDKN2A expression) aggressiveness parameters. Indeed, GOAT overexpression increased, while its silencing/blockade decreased cell-proliferation in PCa cells. Moreover, xenograft tumors derived from GOAT-overexpressing PCa (DU145) cells were significantly higher than those derived from the mock-overexpressing cells. Altogether, our results demonstrate that GOAT could be used as a diagnostic and aggressiveness marker in urine and a therapeutic target in PCa.


2019 ◽  
Vol 113 ◽  
pp. 229-235 ◽  
Author(s):  
Marta Maroto-Diaz ◽  
Natalia Sanz del Olmo ◽  
Laura Muñoz-Moreno ◽  
Ana M. Bajo ◽  
M. José Carmena ◽  
...  

2012 ◽  
Vol 88 (5) ◽  
pp. 405-413 ◽  
Author(s):  
Kinga Brzozowska ◽  
Michael Pinkawa ◽  
Michael J. Eble ◽  
Wolfgang-Ullrich Müller ◽  
Andrzej Wojcik ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (13) ◽  
pp. 3680-3683 ◽  
Author(s):  
R. Jonas A. Nilsson ◽  
Leonora Balaj ◽  
Esther Hulleman ◽  
Sjoerd van Rijn ◽  
D. Michiel Pegtel ◽  
...  

Abstract Diagnostic platforms providing biomarkers that are highly predictive for diagnosing, monitoring, and stratifying cancer patients are key instruments in the development of personalized medicine. We demonstrate that tumor cells transfer (mutant) RNA into blood platelets in vitro and in vivo, and show that blood platelets isolated from glioma and prostate cancer patients contain the cancer-associated RNA biomarkers EGFRvIII and PCA3, respectively. In addition, gene-expression profiling revealed a distinct RNA signature in platelets from glioma patients compared with normal control subjects. Because platelets are easily accessible and isolated, they may form an attractive platform for the companion diagnostics of cancer.


2013 ◽  
Vol 25 (1) ◽  
pp. 245 ◽  
Author(s):  
H.-R. Lee ◽  
S.-H. Hyun ◽  
E.-B. Jeung ◽  
K.-C. Choi

Endocrine-disrupting chemicals (EDC) can bind to the hormone receptor and induce an unexpected hormone response to activate oestrogen receptor (ER)- and androgen receptor (AR)-mediated signalling pathways. Among EDC, bisphenol A (BPA) has a detrimental effect on the endocrine system and is suspected to promote human breast and ovarian cancers. Recent studies have reported that phthalate can disrupt the endocrine system and has a weak estrogenic activity with binding to ER. In this study, we demonstrated whether BPA and dibutyl phthalate (DBP) stimulate the proliferation of prostate cancer cells, LNCaP cells, which have both ER and AR. We evaluated the proliferative rate of LNCaP cells following BPA and DBP treatment using a cell viability assay compared with EtOH treatment as a negative control. Further, we examined the alteration of cell cycle-related gene expressions and TGF-β signalling molecules by semiquantitative RT-PCR. Both BPA and DBP increased LNCaP cell growth more than 2-fold. Moreover, these EDC altered transcriptional expressions of cell cycle-related genes, cyclin D1 and p21, at 6 h in LNCaP cells after exposure of BPA and DBP. Like 17β-oestradiol (E2) and dihydrotestosterone (DHP), treatments of BPA and DBP lead to an increase of the transcriptional levels of c-myc and c-fos in LNCaP cells from 30 min to 6 h. In addition, BPA and DBP decreased the protein level of not only p-smad but also total smad, suggesting that these EDC can affect the molecules of the TGF-β signalling pathway. It was of interest that these effects of EDC were reversed by an antagonist of ER or AR signalling pathways in these prostate cancer cells. These results suggest that BPA and phthalate can alter various gene expressions in TGF-β signalling molecules and stimulate cell growth in prostate cancer cells in vitro. In addition, the growth of prostate cancer cells was stimulated following the exposure of E2, DHT, and DBP in vivo. Taken together, these results indicate the potential of BPA and phthalate in the carcinogenesis of prostate cancer by the oestrogen or androgen-dependent signalling pathway. This work was supported by a National Research Foundation of Korea (NRF) grant funded by the Ministry of Education, Science and Technology (MEST) of Korea government (no. 2011-0015385).


2018 ◽  
Vol 48 (3) ◽  
pp. 1230-1244 ◽  
Author(s):  
Chen Ye ◽  
Chao Zhang ◽  
Hai Huang ◽  
Bo Yang ◽  
Guangan Xiao ◽  
...  

Background/Aims: Natural compounds are a promising resource for anti-tumor drugs. Myricetin, an abundant flavonoid found in the bark and leaves of bayberry, shows multiple promising anti-tumor functions in various cancers. Methods: The cytotoxic, pro-apoptotic, and anti-metastatic effects of myricetin on prostate cancer cells were investigated in both in vitro and in vivo studies. Short-hairpin RNA knockdown of the proviral integration site for Moloney murine leukemia virus-1 (PIM1), pull-down and co-immunoprecipitation assays, and an intracellular Ca2+ flux assay were used to investigate the potential underlying mechanism of myricetin. ONCOMINE database data mining and immunohistochemical analysis of prostate cancer tissues were used to evaluate the expression of PIM1 and CXCR4, as well as the correlation between PIM1 and CXCR4 expression and the clinicopathologic characteristics and prognoses of prostate cancer patients. Results: Myricetin exerted selective cytotoxic, pro-apoptotic, and anti-metastatic effects on prostate cancer cells by inhibiting PIM1 and disrupting the PIM1/CXCR4 interaction. Moreover, PIM1 and CXCR4 were coexpressed and associated with aggressive clinicopathologic traits and poor prognosis in prostate cancer patients. Conclusion: These results offer preclinical evidence for myricetin as a potential chemopreventive and therapeutic agent for precision medicine tailored to prostate cancer patients characterized by concomitant elevated expression of PIM1 and CXCR4.


2021 ◽  
Vol 8 ◽  
Author(s):  
Haotian Tan ◽  
Yaqi Xie ◽  
Xuebao Zhang ◽  
Shuang Wu ◽  
Hongwei Zhao ◽  
...  

Background: Mucosa-associated lymphoma antigen 1 (MALT1) is an oncogene in subsets of diffuse large B cell lymphoma (DLBCL) and mucosa-associated lymphoid tissue type (MALT) lymphoma. However, the role of MALT1 across cancers, especially in prostate cancer is still poorly understood.Methods: Here, we used several public datasets to evaluate MALT1 expression. Then, PCa cell lines and nude mice were used to investigate the cellular functions in vitro and in vivo. Microarray data were downloaded from The Cancer Genome Atlas and MALT1 was subjected to gene set enrichment analysis (GSEA) and Gene Ontology (GO) analysis to identify the biological functions and relevant pathways. Additionally, the correlations between MALT1 expression and mismatch repair (MMR) gene mutation, immune checkpoint gene expression, tumor mutational burden (TMB), and microsatellite instability (MSI) were investigated by Pearson correlation analysis. Moreover, the correlation between MALT1 expression and tumor immune infiltration was analyzed by the Tumor Immune Evaluation Resource (TIMER) database.Results: MALT1 overexpression was significantly correlated with MMR gene mutation levels and crucially promoted proliferation and colony genesis while reducing PCa cell apoptosis levels in vivo and in vitro. MALT1 expression showed strong correlations with immune checkpoint genes, TMB, and MSI in most cancers. The GO analysis indicated that MALT1-coexpressed genes were involved in heterotypic cell-cell adhesion, actin filament-based movement regulation, and action potential regulation. GSEA revealed that MALT1 expression was associated with several signaling pathways, including the NF-κB signaling, Wnt/β-catenin and TGF-β signaling pathways, in PCa. Additionally, MALT1 expression was significantly correlated with the infiltration of immune cells, including B cells, CD8+ T cells, dendritic cells and macrophages, and negatively correlated with CD4+ cell infiltration in PCa.Conclusion: MALT1 expression is higher in pancancer samples than in normal tissues. MALT1 promoted proliferation and colony genesis while reducing PCa cell apoptosis levels, and MALT1 suppression could inhibit xenograft tumor establishment in nude mice. Furthermore, MALT1 expression is closely related to the occurrence and development of multiple tumors in multiple ways. Therefore, MALT1 may be an emerging therapeutic target for a variety of cancers especially PCa.


Sign in / Sign up

Export Citation Format

Share Document