Background

Prostate cancer is the second most frequently diagnosed malignancy and was the fifth leading cause of cancer-related deaths in men worldwide in 2020 [1, 2]. An increasing number of young men have been suffering from prostate cancer [3]. Therefore, the prevention and treatment of prostate cancer are essential. Commonly used treatment methods for prostate cancer include surgical treatment, non-invasive treatment, and androgen therapy [4,5,6]. With emerging treatment methods, such as immunotherapy and molecular therapy [7,8,9], the treatment of prostate cancer and the prognosis have improved. However, prostate cancer prevention and early treatment options are still research hotspots [10].

Acetyl-CoA acetyltransferase 1 (ACAT1) is an important enzyme [11,12,13,14,15,16,3].

In this study, we found that ACAT1 inhibited autophagy in prostate cells and exerted an ROS-scavenging effect. Both autophagy and increased ROS levels have certain tumor-inhibiting effects [27,28,29], and ACAT1 inhibits autophagy and eliminates intracellular ROS mainly by binding to FUS. FUS is an RNA-binding protein, which can promote the production of hnRNA in cells, thereby promoting the transcription of related genes [30]. In this study, we showed that FUS can promote the transcription of LC3B. LC3B is a key protein involved in autophagy. It mainly exists on the membrane of autophagosomes. When LC3A changes to LC3B, autophagosomes are formed. Furthermore, P62 binds to LC3B and autophagosomes on the membrane of a corpuscle; as the autophagosome enters the lysosome, it is degraded [31,32,33]. We believe that autophagy exerts cytotoxicity in prostate cancer. The results of the present study show that after binding to FUS, ACAT1 did not change FUS expression but inhibited FUS from entering the nucleus, thereby inhibiting the production of the autophagy-specific protein LC3B. We speculate that P62 accumulation might be caused by the damage that occurs in the later stages of autophagy [34], which prevents P62 from entering the autophagolysosome, and thereafter, its degradation. Nrf2 is located downstream of P62 [35, 36]. Nrf2 expression increased with the increase in P62 expression, and the active oxygen was scavenged.

Conclusions

Although the mechanism by which ACAT1 promotes prostate cancer has been demonstrated, the mechanism by which ACAT1 prevents FUS from entering the nucleus needs to be further explored. Furthermore, the underlying mechanism of how FUS affects LC3B transcription has not been elucidated and requires further investigation. Nonetheless, we demonstrated that the ACAT1-FUS complex plays a crucial role in prostate cancer development, which provides insights into the development of new therapeutic strategies by targeting or inhibiting the formation of this complex in prostate cancer.