scholarly journals Ketone Body Metabolic Enzyme OXCT1 Regulates Prostate Cancer Chemoresistance

2014 ◽  
Author(s):  
Qiong Liu
Oncogene ◽  
2021 ◽  
Author(s):  
Estefania Labanca ◽  
Juan Bizzotto ◽  
Pablo Sanchis ◽  
Nicolas Anselmino ◽  
Jun Yang ◽  
...  

AbstractProstate cancer (PCa) that progresses after androgen deprivation therapy (ADT) remains incurable. The underlying mechanisms that account for the ultimate emergence of resistance to ADT, progressing to castrate-resistant prostate cancer (CRPC), include those that reactivate androgen receptor (AR), or those that are entirely independent or cooperate with androgen signaling to underlie PCa progression. The intricacy of metabolic pathways associated with PCa progression spurred us to develop a metabolism-centric analysis to assess the metabolic shift occurring in PCa that progresses with low AR expression. We used PCa patient-derived xenografts (PDXs) to assess the metabolic changes after castration of tumor-bearing mice and subsequently confirmed main findings in human donor tumor that progressed after ADT. We found that relapsed tumors had a significant increase in fatty acids and ketone body (KB) content compared with baseline. We confirmed that critical ketolytic enzymes (ACAT1, OXCT1, BDH1) were dysregulated after castrate-resistant progression. Further, these enzymes are increased in the human donor tissue after progressing to ADT. In an in silico approach, increased ACAT1, OXCT1, BDH1 expression was also observed for a subset of PCa patients that relapsed with low AR and ERG (ETS-related gene) expression. Further, expression of these factors was also associated with decreased time to biochemical relapse and decreased progression-free survival. Our studies reveal the key metabolites fueling castration resistant progression in the context of a partial or complete loss of AR dependence.


2016 ◽  
Vol 49 (2) ◽  
pp. 838-846 ◽  
Author(s):  
Jing Li ◽  
Xin Yang ◽  
Hao Guan ◽  
Atsushi Mizokami ◽  
Evan T. Keller ◽  
...  

2021 ◽  
pp. 153535
Author(s):  
Chen Wang ◽  
Tao Ding ◽  
Deping Yang ◽  
Ping Zhang ◽  
Xiongmin Hu ◽  
...  

2017 ◽  
Vol 36 (1) ◽  
Author(s):  
Wenchu Wang ◽  
Lihui Wang ◽  
Atsushi Mizokami ◽  
Junlin Shi ◽  
Chunlin Zou ◽  
...  

2017 ◽  
Author(s):  
Yang Liu ◽  
Liye Xie ◽  
Jing Li ◽  
Atsushi Mizokami ◽  
Evan T. Keller ◽  
...  

2009 ◽  
Vol 101 (7) ◽  
pp. 519-532 ◽  
Author(s):  
Toshiro Migita ◽  
Stacey Ruiz ◽  
Alessandro Fornari ◽  
Michelangelo Fiorentino ◽  
Carmen Priolo ◽  
...  

Biomedicines ◽  
2021 ◽  
Vol 9 (5) ◽  
pp. 538
Author(s):  
Chen Xie ◽  
Pen-Jen Lin ◽  
Jijun Hao

Prostate cancer chemoresistance is a major therapeutic problem, and the underlying mechanism is not well understood and effective therapies to overcome this problem are not available. Phosphodiesterase-4 (PDE4), a main intracellular enzyme for cAMP hydrolysis, has been previously shown to involve in the early chemo-sensitive prostate cancer cell proliferation and progression, but its role in the more-advanced chemo-resistant prostate cancer is completely unknown. Here we found that the expression of PDE4 subtype, PDE4D, is highly elevated in the chemo-resistant prostate cancer cells (DU145-TxR and PC3-TxR) in comparison to the chemo-sensitive prostate cancer cells (DU145 and PC3). Inhibition of PDE4D with a potent and selective PDED4 inhibitor, Eggmanone, effectively decreases the invasion and proliferation as well as induces cell death of the chemo-resistant prostate cancer cells (DU145-TxR and PC3-TxR). These results were confirmed by siRNA knockdown of PDE4D. We and colleagues previously reported that Eggmanone can effectively blocked sonic Hedgehog signaling via PDE4D inhibition, and here our study suggests that that Eggmanone downregulated proliferation of the chemo-resistant prostate cancer cells via sonic Hedgehog signaling. In addition, Eggmanone treatment dose-dependently increases docetaxel cytotoxicity to DU145-TxR and PC3-TxR. As cancer stem cells (CSCs) are known to be implicated in cancer chemoresistance, we further examined Eggmanone impacts on CSC-like properties in the chemo-resistant prostate cancer cells. Our study shows that Eggmanone effectively down-regulates the expression of CSCs’ marker genes Nanog and ABC sub-family G member 2 (ABCG2) and attenuates sphere formation in DU145-TxR and PC3-TxR cells. In summary, our work shows that Eggmanone effectively overcomes the chemoresistance of prostate cancer cells presumably through sonic Hedgehog signaling and targeting CSCs, suggesting that Eggmanone may serve as a novel agent for chemo-resistant prostate cancer.


2020 ◽  
Author(s):  
Kevin Gonthier ◽  
Cindy Weidmann ◽  
Lilianne Frégeau-Proulx ◽  
Étienne Audet-Walsh

2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 11585-11585 ◽  
Author(s):  
Sherri Z. Millis ◽  
Sydney Marsh ◽  
Cristina Pecci ◽  
Sai-Sridhar Boddupalli ◽  
Jeffrey S. Ross ◽  
...  

11585 Background: Fumarate hydratase (FH), an enzyme involved in the Krebs cycle, plays a crucial role in the generation of energy and oxygenation of cells. Genomic alterations (GAs) of FH, a tumor suppressor gene, have been shown to cause chronic hypoxia that encourages tumor formation and have been linked to hereditary leiomyomatosis and renal cell cancer. Only few reports have associated FH mutations with other cancers, and none in prostate cancer. Methods: Identification of an FH V435M pathogenic alteration, which likely changes fumarate binding kinetics, in a prostate cancer patient, with negative family history for renal cancer and cutaneous leiomyomatosis, led to review of a database of 1781 prostate cancer patients, whose tissue was assayed by hybrid-capture based comprehensive genomic profiling (CGP) in the course of clinical care to evaluate genomic alterations (GA: base substitutions, indels, amplifications, copy number alterations, fusions/rearrangements) and targeted therapy opportunities. Tumor mutational burden (TMB) was calculated from a minimum of 1.11 Mb sequenced DNA and reported as mutations/Mb. Results: Profiling identified 49 prostate adenocarcinoma patients (3%) with FH gene alterations, 2 of which harbored the V435M GA identified in the original prostate patient. Ten of 40 alterations were H476_k477 insertions, in the C terminus domain, and 14 were amplifications. The rest were variants of unknown significance (VUS). Conclusions: A FH GA, known to impact other cancers, found in a prostate cancer, led to the discovery of a frequency that suggests deregulation of metabolic pathway activation may contribute to prostate cancer pathogenesis for a subset of patients. The somatic FH GA’s are likely to be substantially more common than germline mutations, and identifying metabolic-enzyme mutations that are pathogenic in prostate cancer could lead to pharmacologic manipulations that are more effective and less toxic than existing therapies. No FDA approved therapies currently exist for this patient’s tumor type nor of any other tumor type with FH GA’s. In our case, alterations in the C-terminal binding domain of FH might inform drug development.


Sign in / Sign up

Export Citation Format

Share Document