AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.7 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

SNORA23 inhibits HCC tumorigenesis by impairing the 2ʹ-O-ribose methylation level of 28S rRNA

Zhiyong Liu1,2,*Yanan Pang3,4,*Yin Jia1Qin Qin1Rui Wang5Wei Li6Jie Jing1Haidong Liu1Shanrong Liu1,7 ( )
Department of Laboratory Diagnostics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
Shanghai Institute of Pancreatic Diseases, Shanghai 200433, China
Department of Chemistry and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433 China
Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200433, China
Shanghai Fourth People’s Hospital, Tongji University School of Medicine, Shanghai 200081, China

*These authors contributed equally to this work.

Show Author Information

Abstract

Objective

The dysregulation of ribosome biogenesis is associated with the progression of numerous tumors, including hepatocellular carcinoma (HCC). Small nucleolar RNAs (snoRNAs) regulate ribosome biogenesis by guiding the modification of ribosomal RNAs (rRNAs). However, the underlying mechanism of this process in HCC remains elusive.

Methods

RNA immunoprecipitation and sequencing were used to analyze RNAs targeted by ribosome proteins. The biological functions of SNORA23 were examined in HCC cells and a xenograft mouse model. To elucidate the underlying mechanisms, the 2ʹ-O-ribose methylation level of rRNAs was evaluated by qPCR, and the key proteins in the PI3K/Akt/mTOR pathway were detected using Western blot.

Results

Twelve snoRNAs were found to co-exist in 4 cancer cell lines using RPS6 pull-down assays. SNORA23 was downregulated in HCC and correlated with the poor prognoses of HCC patients. SNORA23 inhibited the proliferation, migration, and invasion of HCC cells both in vitro and in vivo. We also found that SNORA23 regulated ribosome biogenesis by impairing 2ʹ-O-ribose methylation of cytidine4506 of 28S rRNA. Furthermore, SNORA23, which is regulated by the PI3K/Akt/mTOR signaling pathway, significantly inhibited the phosphorylation of 4E binding protein 1. SNORA23 and rapamycin blocked the PI3K/AKT/mTOR signaling pathway and impaired HCC growth in vivo.

Conclusions

SNORA23 exhibited antitumor effects in HCC and together with rapamycin, provided a promising therapeutic strategy for HCC treatment.

Electronic Supplementary Material

Download File(s)
cbm-19-1-104_ESM.pdf (973.1 KB)

References

1

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA-Cancer J Clin. 2019; 69: 7-34.

2

Wang X, Liao X, Yang C, Huang K, Yu T, Yu L, et al. Identification of prognostic biomarkers for patients with hepatocellular carcinoma after hepatectomy. Oncol Rep. 2019; 41: 1586-602.

3

Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastro Hepat. 2019; 16: 589-604.

4

Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in china: good or bad news from the 2018 global cancer statistics? Cancer Commun (London, England). 2019; 39: 22.

5

Xu RH, Wei W, Krawczyk M, Wang W, Luo H, Flagg K, et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater. 2017; 16: 1155-61.

6

Pelletier J, Thomas G, Volarević S. Ribosome biogenesis in cancer: new players and therapeutic avenues. Nat Rev Cancer. 2018; 18: 51-63.

7

Bustelo XR, Dosil M. Ribosome biogenesis and cancer: basic and translational challenges. Curr Opin Genet Dev. 2018; 48: 22-9.

8

Turi Z, Lacey M, Mistrik M, Moudry P. Impaired ribosome biogenesis: mechanisms and relevance to cancer and aging. Aging. 2019; 11: 2512-40.

9

Devlin JR, Hannan KM, Hein N, Cullinane C, Kusnadi E, Ng PY, et al. Combination therapy targeting ribosome biogenesis and mRNA translation synergistically extends survival in MYC-driven lymphoma. Cancer Discov. 2016; 6: 59-70.

10

van Riggelen J, Yetil A, Felsher DW. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat Rev Cancer. 2010; 10: 301-9.

11

Morcelle C, Menoyo S, Morón-Duran FD, Tauler A, Kozma SC, Thomas G, et al. Oncogenic MYC induces the impaired ribosome biogenesis checkpoint and stabilizes p53 independent of increased ribosome content. Cancer Res. 2019; 79: 4348-59.

12

Williams GT, Farzaneh F. Are snornas and snorna host genes new players in cancer? Nature reviews. Cancer. 2012; 12: 84-8.

13

Kim DS, Camacho CV, Nagari A, Malladi VS, Challa S, Kraus WL. Activation of PARP-1 by snoRNAs controls ribosome biogenesis and cell growth via the RNA helicase DDX21. Mol Cell. 2019; 75: 1270-85.

14

McMahon M, Contreras A, Holm M, Uechi T, Forester CM, Pang X, et al. A single H/ACA small nucleolar RNA mediates tumor suppression downstream of oncogenic RAS. eLife. 2019; 8: e48847.

15

Liang J, Wen J, Huang Z, Chen XP, Zhang BX, Chu L. Small nucleolar RNAs: insight into their function in cancer. Front Oncol. 2019; 9: 587.

16

Baral D, Wu L, Katwal G, Yan X, Wang Y, Ye Q. Clinical significance and biological roles of small nucleolar RNAs in hepatocellular carcinoma. Bio Rep. 2018; 8: 319-24.

17

Wu L, Chang L, Wang H, Ma W, Peng Q, Yuan Y. Clinical significance of C/D box small nucleolar RNA U76 as an oncogene and a prognostic biomarker in hepatocellular carcinoma. Clin Res Hepatol Gas. 2018; 42: 82-91.

18

Shuwen H, Xi Y, Quan Q, Yin J, Miao D. Can small nucleolar RNA be a novel molecular target for hepatocellular carcinoma? Gene. 2020; 733: 144384.

19

Rahmani F, Ziaeemehr A, Shahidsales S, Gharib M, Khazaei M, Ferns GA, et al. Role of regulatory miRNAs of the PI3k/AKT/mTOR signaling in the pathogenesis of hepatocellular carcinoma. J Cell Physiol. 2020; 235: 4146-52.

20

Wang C, Cigliano A, Jiang L, Li X, Fan B, Pilo MG, et al. 4EBP1/eIF4E and p70s6k/RPs6 axes play critical and distinct roles in hepatocarcinogenesis driven by AKT and N-Ras proto-oncogenes in mice. Hepatology (Baltimore, Md.). 2015; 61: 200-13.

21

Penzo M, Montanaro L, Treré D, Derenzini M. The ribosome biogenesis-cancer connection. Cell. 2019; 8: 55.

22

Dupuis-Sandoval F, Poirier M, Scott MS. The emerging landscape of small nucleolar RNAs in cell biology. Wiley interdisciplinary reviews. RNA. 2015; 6: 381-97.

23

Meyuhas O. Ribosomal protein S6 phosphorylation: four decades of research. Int Rev Cell Mol Bio. 2015; 320: 41-73.

24

Golob-Schwarzl N, Krassnig S, Toeglhofer AM, Park YN, Gogg-Kamerer M, Vierlinger K, et al. New liver cancer biomarkers: PI3k/AKT/mTOR pathway members and eukaryotic translation initiation factors. Eur J Cancer (Oxford, England: 1990). 2017; 83: 56-70.

25

Han S, Khuri FR, Roman J. Fibronectin stimulates non-small cell lung carcinoma cell growth through activation of Akt/mammalian target of rapamycin/S6 kinase and inactivation of LKB1/AMP-activated protein kinase signal pathways. Cancer Res. 2006; 66: 315-23.

26

Gong J, Li Y, Liu CJ, Xiang Y, Li C, Ye Y, et al. A pan-cancer analysis of the expression and clinical relevance of small nucleolar rnas in human cancer. Cell Rep. 2017; 21: 1968-81.

27

Kiss AM, Jády BE, Bertrand E, Kiss T. Human box H/ACA pseudouridylation guide RNA machinery. Mol Cell Biol. 2004; 24: 5797-807.

28

Yoshihama M, Nakao A, Kenmochi N. snOPY: a small nucleolar RNA orthological gene database. BMC Res Not. 2013; 6: 426.

29

Jády BE, Kiss T. A small nucleolar guide RNA functions both in 2’-o-ribose methylation and pseudouridylation of the U5 spliceosomal RNA. EMBO J. 2001; 20: 541-51.

30

Carotenuto P, Pecoraro A, Palma G, Russo G, Russo A. Therapeutic approaches targeting nucleolus in cancer. Cell. 2019; 8: 1090.

31

Biedka S, Micic J, Wilson D, Brown H, Diorio-Toth L, Woolford JL, Jr. Hierarchical recruitment of ribosomal proteins and assembly factors remodels nucleolar pre-60S ribosomes. J Cell Biol. 2018; 217: 2503-18.

32

Cao P, Yang A, Wang R, Xia X, Zhai Y, Li Y, et al. Germline duplication of SNORA18l5 increases risk for HBV-related hepatocellular carcinoma by altering localization of ribosomal proteins and decreasing levels of p53. Gastroenterology. 2018; 155: 542-56.

33

Sloan KE, Warda AS, Sharma S, Entian KD, Lafontaine DLJ, Bohnsack MT. Tuning the ribosome: The influence of rRNA modification on eukaryotic ribosome biogenesis and function. RNA Biol. 2017; 14: 1138-52.

34

Cui L, Nakano K, Obchoei S, Setoguchi K, Matsumoto M, Yamamoto T, et al. Small nucleolar noncoding RNA SNORA23, up-regulated in human pancreatic ductal adenocarcinoma, regulates expression of spectrin repeat-containing nuclear envelope 2 to promote growth and metastasis of xenograft tumors in mice. Gastroenterology. 2017; 153: 292-306.

35

Iyer-Bierhoff A, Krogh N, Tessarz P, Ruppert T, Nielsen H, Grummt I. SIRT7-dependent deacetylation of fibrillarin controls histone H2a methylation and rRNA synthesis during the cell cycle. Cell Rep. 2018; 25: 2946-54.

36

Marcel V, Ghayad SE, Belin S, Therizols G, Morel AP, Solano-Gonzàlez E, et al. P53 acts as a safeguard of translational control by regulating fibrillarin and rRNA methylation in cancer. Cancer Cell. 2013; 24: 318-30.

37

Baudin-Baillieu A, Fabret C, Liang XH, Piekna-Przybylska D, Fournier MJ, Rousset JP. Nucleotide modifications in three functionally important regions of the saccharomyces cerevisiae ribosome affect translation accuracy. Nucleic Acids Res. 2009; 37: 7665-77.

38

Zhou F, Liu Y, Rohde C, Pauli C, Gerloff D, Köhn M, et al. AML1-ETO requires enhanced C/D box snoRNA/RNP formation to induce self-renewal and leukaemia. Nat Cell Biol. 2017; 19: 844-55.

39

Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, et al. Oncogenic signaling pathways in the cancer genome atlas. Cell. 2018; 173: 321-37.

40

Nusse R, Clevers H. Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 2017; 169: 985-99.

41

Guertin DA, Sabatini DM. The pharmacology of mtor inhibition. Sci Signal. 2009; 2: 24.

42

Choo AY, Yoon SO, Kim SG, Roux PP, Blenis J. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci USA. 2008; 105: 17414-9.

Cancer Biology & Medicine
Pages 104-119
Cite this article:
Liu Z, Pang Y, Jia Y, et al. SNORA23 inhibits HCC tumorigenesis by impairing the 2ʹ-O-ribose methylation level of 28S rRNA. Cancer Biology & Medicine, 2022, 19(1): 104-119. https://doi.org/10.20892/j.issn.2095-3941.2020.0343

173

Views

1

Downloads

7

Crossref

0

Web of Science

13

Scopus

Altmetrics

Received: 28 June 2020
Accepted: 11 November 2020
Published: 21 March 2022
©2022 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return