MiRNAs and snoRNAs in Bone Metastasis: Functional Roles and Clinical Potential
Abstract
:Simple Summary
Abstract
1. Introduction
2. Current Understanding of Bone Metastasis
3. MiRNAs and SnoRNAs: Small yet Important Non-Coding RNAs
3.1. Biogenesis and Biological Functions of MiRNAs
3.2. Biogenesis and Biological Functions of SnoRNAs
4. Role of Circulating MiRNAs and SnoRNAs in Bone Metastasis
4.1. MiRNA and SnoRNA Roles in the Formation of a Pre-Metastatic Niche
4.2. MiRNA and SnoRNA Roles in the Vicious Cycle in Bone
5. MiRNA and SnoRNA Expression in Cancer Cells and Their Roles in Bone Metastasis Progression
6. Circulating MiRNAs and SnoRNA as Potential Biomarkers
6.1. Use of Circulating MiRNAs and SnoRNAs as Biomarkers: Some Examples
6.2. Technical Strengths and Limitations of MiRNA and SnoRNA Biomarkers
7. Therapeutic Opportunities from MiRNAs and SnoRNA Use
8. Conclusions and Future Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
ASO | antisense oligonucleotide |
BMSC | bone marrow mesenchymal stem cells |
BMP | bone morphogenetic protein |
COL1A1 | collagenase 1-A1 |
EMT | epithelial-to-mesenchymal transition |
EV | extracellular vesicle |
FBL | fibrillarin |
IBSP | integrin-binding sialoprotein |
IL | interleukin |
IRES | internal ribosome entry site |
LOX | hypoxia-induced lysyl oxidase |
lncRNA | long non-coding RNAs |
M-CSF | macrophage-colony stimulating factor |
MET | mesenchymal-to-epithelial transition |
miRNA | microRNA |
NGS | next-generation sequencing |
ncRNA | non-coding RNA |
PGF | placental growth factor |
PSA | prostate-specific antigen |
PRDX-4 | peroxiredoxin-4 |
PTHrP | type 1 parathyroid hormone |
RANKL | receptor activator of nuclear factor kappa-beta ligand |
rRNA | ribosomal RNA |
RUNX2 | runt-related transcription factor 2 |
scaRNA | small Cajal body-associated snoRNA |
sncRNA | small non-coding RNA |
snoRA | box H/ACA snoRNA |
snoRD | box C/D snoRNA |
snoRNA | small nucleolar RNA |
snoRNP | small nucleolar ribonucleoprotein |
tRNA | transfer RNA |
TGF-β | transforming growth factor-beta |
TGIRT-seq | thermostable group II intron reverse transcriptase sequencing |
VEGF-A | vascular endothelial growth factor A |
Wnt | Wingless-INT |
References
- Chaffer, C.L.; San Juan, B.P.; Lim, E.; Weinberg, R.A. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 2016, 35, 645–654. [Google Scholar] [CrossRef]
- Clézardin, P.; Coleman, R.; Puppo, M.; Ottewell, P.; Bonnelye, E.; Paycha, F.; Confavreux, C.B.; Holen, I. Bone metastasis: Mechanisms, therapies, and biomarkers. Physiol. Rev. 2021, 101, 797–855. [Google Scholar] [CrossRef]
- Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
- Coleman, R.E.; Croucher, P.I.; Padhani, A.R.; Clézardin, P.; Chow, E.; Fallon, M.; Guise, T.; Colangeli, S.; Capanna, R.; Costa, L. Bone metastases. Nat. Rev. Dis. Primers 2020, 6, 83. [Google Scholar] [CrossRef]
- Palazzo, A.F.; Lee, E.S. Non-coding RNA: What is functional and what is junk? Front. Genet. 2015, 6, 2. [Google Scholar] [CrossRef] [Green Version]
- Puppo, M.; Taipaleenmäki, H.; Hesse, E.; Clézardin, P. Non-coding RNAs in bone remodelling and bone metastasis: Mechanisms of action and translational relevance. Br. J. Pharmacol. 2021, 178, 1936–1954. [Google Scholar] [CrossRef]
- Anastasiadou, E.; Jacob, L.S.; Slack, F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer 2018, 18, 5–18. [Google Scholar] [CrossRef]
- Puppo, M.; Valluru, M.K.; Clézardin, P. Chapter 33—MicroRNAs and bone metastasis: How small RNAs regulate secondary tumor formation and progression in the skeleton. In Bone Sarcomas and Bone Metastases—From Bench to Bedside, 3rd ed.; Heymann, D., Ed.; Academic Press: Cambridge, MA, USA, 2022; pp. 457–469. [Google Scholar]
- Macedo, F.; Ladeira, K.; Pinho, F.; Saraiva, N.; Bonito, N.; Pinto, L.; Gonçalves, F. Bone metastases: An overview. Oncol. Rev. 2017, 11, 321. [Google Scholar]
- Coleman, R.E. Clinical Features of Metastatic Bone Disease and Risk of Skeletal Morbidity. Clin. Cancer Res. 2006, 12, 6243s–6249s. [Google Scholar] [CrossRef] [Green Version]
- Berardi, R.; Berruti, A.; Brogelli, L.; Zucali, P.A. An Italian survey on the use of denosumab for the management of skeletal-related events in patients with bone metastases. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 4659–4665. [Google Scholar]
- Le Pape, F.; Vargas, G.; Clézardin, P. The role of osteoclasts in breast cancer bone metastasis. J. Bone Oncol. 2016, 5, 93–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suva, L.J.; Washam, C.; Nicholas, R.W.; Griffin, R.J. Bone metastasis: Mechanisms and therapeutic opportunities. Nat. Rev. Endocrinol. 2011, 7, 208–218. [Google Scholar] [CrossRef] [PubMed]
- Katagiri, T.; Yamaguchi, A.; Komaki, M.; Abe, E.; Takahashi, N.; Ikeda, T.; Rosen, V.; Wozney, J.M.; Fujisawa-Sehara, A.; Suda, T. Bone morphogenetic protein-2 converts the differentiation pathway of C2C12 myoblasts into the osteoblast lineage. J. Cell Biol. 1994, 127 Pt 1, 1755–1766. [Google Scholar] [CrossRef] [Green Version]
- Janssens, K.; ten Dijke, P.; Janssens, S.; Van Hul, W. Transforming growth factor-beta1 to the bone. Endocr. Rev. 2005, 26, 743–774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, W.; Bado, I.; Wang, H.; Lo, H.-C.; Zhang, X.H.-F. Bone Metastasis: Find Your Niche and Fit in. Trends Cancer 2019, 5, 95–110. [Google Scholar] [CrossRef]
- Mundy, G.R. Mechanisms of bone metastasis. Cancer 1997, 80 (Suppl 8), 1546–1556. [Google Scholar] [CrossRef]
- Riquelme, M.A.; Cardenas, E.R.; Jiang, J.X. Osteocytes and Bone Metastasis. Front. Endocrinol. 2020, 11, 567844. [Google Scholar] [CrossRef]
- He, N.; Jiang, J. Contribution of immune cells to bone metastasis pathogenesis. Front. Endocrinol. 2022, 13, 1019864. [Google Scholar] [CrossRef]
- Mukaida, N.; Zhang, D.; Sasaki, S.-I. Emergence of Cancer-Associated Fibroblasts as an Indispensable Cellular Player in Bone Metastasis Process. Cancers 2020, 12, 2896. [Google Scholar] [CrossRef]
- Weilbaecher, K.N.; Guise, T.A.; McCauley, L.K. Cancer to bone: A fatal attraction. Nat. Rev. Cancer 2011, 11, 411–425. [Google Scholar] [CrossRef] [Green Version]
- Luzzi, K.J.; Macdonald, I.C.; Schmidt, E.E.; Kerkvliet, N.; Morris, V.L.; Chambers, A.F.; Groom, A.C. Multistep Nature of Metastatic Inefficiency: Dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am. J. Pathol. 1998, 153, 865–873. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef] [PubMed]
- Holen, I.; Lefley, D.V.; Francis, S.E.; Rennicks, S.; Bradbury, S.; Coleman, R.E.; Ottewell, P. IL-1 drives breast cancer growth and bone metastasis in vivo. Oncotarget 2016, 7, 75571–75584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cox, T.R.; Rumney, R.M.H.; Schoof, E.M.; Perryman, L.; Høye, A.M.; Agrawal, A.; Bird, D.; Ab Latif, N.; Forrest, H.; Evans, H.R.; et al. The hypoxic cancer secretome induces pre-metastatic bone lesions through lysyl oxidase. Nature 2015, 522, 106–110. [Google Scholar] [CrossRef] [Green Version]
- Puppo, M.; Valluru, M.K.; Clézardin, P. MicroRNAs and Their Roles in Breast Cancer Bone Metastasis. Curr. Osteoporos. Rep. 2021, 19, 256–263. [Google Scholar] [CrossRef]
- Jaafar, M.; Paraqindes, H.; Gabut, M.; Diaz, J.J.; Marcel, V.; Durand, S. 2’O-Ribose Methylation of Ribosomal RNAs: Natural Diversity in Living Organisms, Biological Processes, and Diseases. Cells 2021, 10, 1948. [Google Scholar] [CrossRef]
- Samarsky, D.A.; Fournier, M.J.; Singer, R.H.; Bertrand, E. The snoRNA box C/D motif directs nucleolar targeting and also couples snoRNA synthesis and localization. EMBO J. 1998, 17, 3747–3757. [Google Scholar] [CrossRef]
- van der Werf, J.; Chin, C.V.; Fleming, N.I. SnoRNA in Cancer Progression, Metastasis and Immunotherapy Response. Biology 2021, 10, 809. [Google Scholar] [CrossRef]
- Brameier, M.; Herwig, A.; Reinhardt, R.; Walter, L.; Gruber, J. Human box C/D snoRNAs with miRNA like functions: Expanding the range of regulatory RNAs. Nucleic Acids Res. 2011, 39, 675–686. [Google Scholar] [CrossRef]
- Duchaine, T.F.; Fabian, M.R. Mechanistic Insights into MicroRNA-Mediated Gene Silencing. Cold Spring Harb. Perspect. Biol. 2019, 11, a032771. [Google Scholar] [CrossRef] [Green Version]
- Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vasudevan, S.; Tong, Y.; Steitz, J.A. Switching from Repression to Activation: MicroRNAs Can Up-Regulate Translation. Science 2007, 318, 1931–1934. [Google Scholar] [CrossRef] [PubMed]
- Biasini, A.; Abdulkarim, B.; de Pretis, S.; Tan, J.Y.; Arora, R.; Wischnewski, H.; Dreos, R.; Pelizzola, M.; Ciaudo, C.; Marques, A.C. Translation is required for miRNA-dependent decay of endogenous transcripts. EMBO J. 2021, 40, e104569. [Google Scholar] [CrossRef] [PubMed]
- Weidle, U.H.; Nopora, A. Long Non-coding RNAs Sponging MicroRNAs with Efficacy in Preclinical In Vivo Models of Esophageal Squamous Cell Cancer. Anticancer Res. 2022, 42, 3233. [Google Scholar] [CrossRef] [PubMed]
- Maxwell, E.S.; Fournier, M.J. The small nucleolar RNAs. Annu. Rev. Biochem. 1995, 64, 897–934. [Google Scholar] [CrossRef]
- Huang, Z.H.; Du, Y.P.; Wen, J.T.; Lu, B.F.; Zhao, Y. snoRNAs: Functions and mechanisms in biological processes, and roles in tumor pathophysiology. Cell Death Discov. 2022, 8, 259. [Google Scholar] [CrossRef]
- Tollervey, D.; Kiss, T. Function and synthesis of small nucleolar RNAs. Curr. Opin. Cell Biol. 1997, 9, 337–342. [Google Scholar] [CrossRef]
- Vitali, P.; Kiss, T. Cooperative 2’-O-methylation of the wobble cytidine of human elongator tRNA(Met)(CAT) by a nucleolar and a Cajal body-specific box C/D RNP. Genes Dev. 2019, 33, 741–746. [Google Scholar] [CrossRef] [Green Version]
- Jorjani, H.; Kehr, S.; Jedlinski, D.J.; Gumienny, R.; Hertel, J.; Stadler, P.F.; Zavolan, M.; Gruber, A.R. An updated human snoRNAome. Nucleic Acids Res. 2016, 44, 5068–5082. [Google Scholar] [CrossRef]
- Ojha, S.; Malla, S.; Lyons, S.M. snoRNPs: Functions in Ribosome Biogenesis. Biomolecules 2020, 10, 783. [Google Scholar] [CrossRef]
- Marcel, V.; Ghayad, S.E.; Belin, S.; Therizols, G.; Morel, A.-P.; Solano-Gonzàlez, E.; Vendrell, J.A.; Hacot, S.; Mertani, H.C.; Albaret, M.A.; et al. p53 Acts as a Safeguard of Translational Control by Regulating Fibrillarin and rRNA Methylation in Cancer. Cancer Cell 2013, 24, 318–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Erales, J.; Marchand, V.; Panthu, B.; Gillot, S.; Belin, S.; Ghayad, S.E.; Garcia, M.; Laforêts, F.; Marcel, V.; Baudin-Baillieu, A.; et al. Evidence for rRNA 2′-O-methylation plasticity: Control of intrinsic translational capabilities of human ribosomes. Proc. Natl. Acad. Sci. USA 2017, 114, 12934–12939. [Google Scholar] [CrossRef] [PubMed]
- Mourksi, N.-E.; Morin, C.; Fenouil, T.; Diaz, J.-J.; Marcel, V. snoRNAs Offer Novel Insight and Promising Perspectives for Lung Cancer Understanding and Management. Cells 2020, 9, 541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barros-Silva, D.; Klavert, J.; Jenster, G.; Jerónimo, C.; Lafontaine, D.L.; Martens-Uzunova, E.S. The role of OncoSnoRNAs and Ribosomal RNA 2’-O-methylation in Cancer. RNA Biol. 2021, 18 (Suppl 1), 61–74. [Google Scholar] [CrossRef]
- Dsouza, V.L.; Adiga, D.; Sriharikrishnaa, S.; Suresh, P.S.; Chatterjee, A.; Kabekkodu, S.P. Small nucleolar RNA and its potential role in breast cancer—A comprehensive review. Biochim. Biophys. Acta 2021, 1875, 188501. [Google Scholar] [CrossRef]
- Sharma, S.; Yang, J.; van Nues, R.; Watzinger, P.; Kötter, P.; Lafontaine, D.L.J.; Granneman, S.; Entian, K.-D. Specialized box C/D snoRNPs act as antisense guides to target RNA base acetylation. PLoS Genet. 2017, 13, e1006804. [Google Scholar] [CrossRef] [Green Version]
- Dudnakova, T.; Dunn-Davies, H.; Peters, R.; Tollervey, D. Mapping targets for small nucleolar RNAs in yeast. Wellcome Open Res. 2018, 3, 120. [Google Scholar] [CrossRef]
- O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. [Google Scholar] [CrossRef]
- Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.-J.; Xie, G.-Y.; Miao, Y.-R.; Xia, M.; Wang, Y.; Lei, Q.; Zhang, Q.; Guo, A.-Y. EVAtlas: A comprehensive database for ncRNA expression in human extracellular vesicles. Nucleic Acids Res. 2022, 50, D111–D117. [Google Scholar] [CrossRef]
- Zimta, A.-A.; Sigurjonsson, O.E.; Gulei, D.; Tomuleasa, C. The Malignant Role of Exosomes as Nanocarriers of Rare RNA Species. Int. J. Mol. Sci. 2020, 21, 5866. [Google Scholar] [CrossRef] [PubMed]
- Lapidot, T.; Petit, I. Current understanding of stem cell mobilization: The roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Exp. Hematol. 2002, 30, 973–981. [Google Scholar] [CrossRef] [PubMed]
- Massagué, J.; Obenauf, A.C. Metastatic colonization by circulating tumour cells. Nature 2016, 529, 298–306. [Google Scholar] [CrossRef] [Green Version]
- Raimondi, L.; De Luca, A.; Amodio, N.; Manno, M.; Raccosta, S.; Taverna, S.; Bellavia, D.; Naselli, F.; Fontana, S.; Schillaci, O.; et al. Involvement of multiple myeloma cell-derived exosomes in osteoclast differentiation. Oncotarget 2015, 6, 13772–13789. [Google Scholar] [CrossRef] [Green Version]
- Karlsson, T.; Lundholm, M.; Widmark, A.; Persson, E. Tumor Cell-Derived Exosomes from the Prostate Cancer Cell Line TRAMP-C1 Impair Osteoclast Formation and Differentiation. PLoS ONE 2016, 11, e0166284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dai, J.; Escara-Wilke, J.; Keller, J.M.; Jung, Y.; Taichman, R.S.; Pienta, K.J.; Keller, E.T. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J. Exp. Med. 2019, 216, 2883–2899. [Google Scholar] [CrossRef] [PubMed]
- Probert, C.; Dottorini, T.; Speakman, A.; Hunt, S.; Nafee, T.; Fazeli, A.; Wood, S.; Brown, J.E.; James, V. Communication of prostate cancer cells with bone cells via extracellular vesicle RNA; a potential mechanism of metastasis. Oncogene 2019, 38, 1751–1763. [Google Scholar] [CrossRef]
- Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R.; et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [Green Version]
- Le, M.T.; Hamar, P.; Guo, C.; Basar, E.; Perdigão-Henriques, R.; Balaj, L.; Lieberman, J. miR-200–containing extracellular vesicles promote breast cancer cell metastasis. J. Clin. Investig. 2014, 124, 5109–5128. [Google Scholar] [CrossRef] [Green Version]
- Guo, L.; Zhu, Y.; Li, L.; Zhou, S.; Yin, G.; Yu, G.; Cui, H. Breast cancer cell-derived exosomal miR-20a-5p promotes the proliferation and differentiation of osteoclasts by targeting SRCIN1. Cancer Med. 2019, 8, 5687–5701. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Wu, J. The Potential Roles of Exosomal miR-214 in Bone Metastasis of Lung Adenocarcinoma. Front. Oncol. 2020, 10, 611054. [Google Scholar] [CrossRef] [PubMed]
- Ye, Y.; Li, S.-L.; Ma, Y.-Y.; Diao, Y.-J.; Yang, L.; Su, M.-Q.; Li, Z.; Ji, Y.; Wang, J.; Lei, L.; et al. Exosomal miR-141-3p regulates osteoblast activity to promote the osteoblastic metastasis of prostate cancer. Oncotarget 2017, 8, 94834–94849. [Google Scholar] [CrossRef] [PubMed]
- Wu, K.; Feng, J.; Lyu, F.; Xing, F.; Sharma, S.; Liu, Y.; Wu, S.-Y.; Zhao, D.; Tyagi, A.; Deshpande, R.P.; et al. Exosomal miR-19a and IBSP cooperate to induce osteolytic bone metastasis of estrogen receptor-positive breast cancer. Nat. Commun. 2021, 12, 5196. [Google Scholar] [CrossRef] [PubMed]
- Hashimoto, K.; Ochi, H.; Sunamura, S.; Kosaka, N.; Mabuchi, Y.; Fukuda, T.; Yao, K.; Kanda, H.; Ae, K.; Okawa, A.; et al. Cancer-secreted hsa-miR-940 induces an osteoblastic phenotype in the bone metastatic microenvironment via targeting ARHGAP1 and FAM134A. Proc. Natl. Acad. Sci. USA 2018, 115, 2204–2209. [Google Scholar] [CrossRef] [Green Version]
- Xu, H.; Yao, J.; Wu, D.C.; Lambowitz, A.M. Improved TGIRT-seq methods for comprehensive transcriptome profiling with decreased adapter dimer formation and bias correction. Sci. Rep. 2019, 9, 7953. [Google Scholar] [CrossRef] [Green Version]
- Rai, A.K.; Rajan, K.S.; Bisserier, M.; Brojakowska, A.; Sebastian, A.; Evans, A.C.; Coleman, M.A.; Mills, P.J.; Arakelyan, A.; Uchida, S.; et al. Spaceflight-Associated Changes of snoRNAs in Peripheral Blood Mononuclear Cells and Plasma Exosomes—A Pilot Study. Front. Cardiovasc. Med. 2022, 9, 886689. [Google Scholar] [CrossRef]
- Furesi, G.; Domingues, A.M.D.J.; Alexopoulou, D.; Dahl, A.; Hackl, M.; Schmidt, J.R.; Kalkhof, S.; Kurth, T.; Taipaleenmäki, H.; Conrad, S.; et al. Exosomal miRNAs from Prostate Cancer Impair Osteoblast Function in Mice. Int. J. Mol. Sci. 2022, 23, 1285. [Google Scholar] [CrossRef]
- Yu, L.; Sui, B.; Fan, W.; Lei, L.; Zhou, L.; Yang, L.; Diao, Y.; Zhang, Y.; Li, Z.; Liu, J.; et al. Exosomes derived from osteogenic tumor activate osteoclast differentiation and concurrently inhibit osteogenesis by transferring COL1A1-targeting miRNA-92a-1-5p. J. Extracell. Vesicles 2021, 10, e12056. [Google Scholar] [CrossRef]
- Grigsby, I.F.; Pham, L.; Gopalakrishnan, R.; Mansky, L.M.; Mansky, K.C. Downregulation of Gnas, Got2 and Snord32a following tenofovir exposure of primary osteoclasts. Biochem. Biophys. Res. Commun. 2010, 391, 1324–1329. [Google Scholar] [CrossRef] [Green Version]
- Khor, E.-C.; Fanshawe, B.; Qi, Y.; Zolotukhin, S.; Kulkarni, R.N.; Enriquez, R.F.; Purtell, L.; Lee, N.J.; Wee, N.K.; Croucher, P.I.; et al. Prader-Willi Critical Region, a Non-Translated, Imprinted Central Regulator of Bone Mass: Possible Role in Skeletal Abnormalities in Prader-Willi Syndrome. PLoS ONE 2016, 11, e0148155. [Google Scholar] [CrossRef] [Green Version]
- Qi, Y.; Purtell, L.; Fu, M.; Sengmany, K.; Loh, K.; Zhang, L.; Zolotukhin, S.; Sainsbury, A.; Campbell, L.; Herzog, H. Ambient temperature modulates the effects of the Prader-Willi syndrome candidate gene Snord116 on energy homeostasis. Neuropeptides 2017, 61, 87–93. [Google Scholar] [CrossRef] [PubMed]
- Steinbusch, M.M.F.; Caron, M.M.J.; Surtel, D.A.M.; Friedrich, F.; Lausch, E.; Pruijn, G.J.M.; Verhesen, W.; Schroen, B.L.M.; van Rhijn, L.W.; Zabel, B.; et al. Expression of RMRP RNA is regulated in chondrocyte hypertrophy and determines chondrogenic differentiation. Sci. Rep. 2017, 7, 6440. [Google Scholar] [CrossRef]
- Pourebrahim, R.; Zhang, Y.; Liu, B.; Gao, R.; Xiong, S.; Lin, P.P.; McArthur, M.J.; Ostrowski, M.C.; Lozano, G. Integrative genome analysis of somatic p53 mutant osteosarcomas identifies Ets2-dependent regulation of small nucleolar RNAs by mutant p53 protein. Genes Dev. 2017, 31, 1847–1857. [Google Scholar] [CrossRef] [PubMed]
- Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Mark, M.T.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tiedemann, K.; Sadvakassova, G.; Mikolajewicz, N.; Juhas, M.; Sabirova, Z.; Tabariès, S.; Gettemans, J.; Siegel, P.M.; Komarova, S.V. Exosomal Release of L-Plastin by Breast Cancer Cells Facilitates Metastatic Bone Osteolysis. Transl. Oncol. 2019, 12, 462–474. [Google Scholar] [CrossRef]
- Soe, Z.; Park, E.; Shimaoka, M. Integrin Regulation in Immunological and Cancerous Cells and Exosomes. Int. J. Mol. Sci. 2021, 22, 2193. [Google Scholar] [CrossRef]
- Ma, L.; Teruya-Feldstein, J.; Weinberg, R.A. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 2007, 449, 682–688. [Google Scholar] [CrossRef]
- Haider, M.-T.; Smit, D.J.; Taipaleenmäki, H. MicroRNAs: Emerging Regulators of Metastatic Bone Disease in Breast Cancer. Cancers 2022, 14, 729. [Google Scholar] [CrossRef]
- Oh-Hohenhorst, S.J.; Lange, T. Role of Metastasis-Related microRNAs in Prostate Cancer Progression and Treatment. Cancers 2021, 13, 4492. [Google Scholar] [CrossRef]
- Wu, S.-G.; Chang, T.-H.; Liu, Y.-N.; Shih, J.-Y. MicroRNA in Lung Cancer Metastasis. Cancers 2019, 11, 265. [Google Scholar] [CrossRef] [Green Version]
- Kinget, L.; Roussel, E.; Lambrechts, D.; Boeckx, B.; Vanginderhuysen, L.; Albersen, M.; Rodríguez-Antona, C.; Graña-Castro, O.; Inglada-Pérez, L.; Verbiest, A.; et al. MicroRNAs Possibly Involved in the Development of Bone Metastasis in Clear-Cell Renal Cell Carcinoma. Cancers 2021, 13, 1554. [Google Scholar] [CrossRef] [PubMed]
- Gajos-Michniewicz, A.; Czyz, M. Role of miRNAs in Melanoma Metastasis. Cancers 2019, 11, 326. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, V.H.L.; Yue, C.; Du, K.Y.; Salem, M.; O’Brien, J.; Peng, C. The Role of microRNAs in Epithelial Ovarian Cancer Metastasis. Int. J. Mol. Sci. 2020, 21, 7093. [Google Scholar] [CrossRef] [PubMed]
- Celano, M.; Rosignolo, F.; Maggisano, V.; Pecce, V.; Iannone, M.; Russo, D.; Bulotta, S. MicroRNAs as Biomarkers in Thyroid Carcinoma. Int. J. Genom. 2017, 2017, 6496570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhuo, Y.; Li, S.; Hu, W.; Zhang, Y.; Shi, Y.; Zhang, F.; Zhang, J.; Wang, J.; Liao, M.; Chen, J.; et al. Targeting SNORA38B attenuates tumorigenesis and sensitizes immune checkpoint blockade in non-small cell lung cancer by remodeling the tumor microenvironment via regulation of GAB2/AKT/mTOR signaling pathway. J. Immunother. Cancer 2022, 10, e004113. [Google Scholar] [CrossRef] [PubMed]
- Cui, L.; Nakano, K.; Obchoei, S.; Setoguchi, K.; Matsumoto, M.; Yamamoto, T.; Obika, S.; Shimada, K.; Hiraoka, N. 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.e2. [Google Scholar] [CrossRef]
- Yi, C.; Wan, X.; Zhang, Y.; Fu, F.; Zhao, C.; Qin, R.; Wu, H.; Li, Y.; Huang, Y. SNORA42 enhances prostate cancer cell viability, migration and EMT and is correlated with prostate cancer poor prognosis. Int. J. Biochem. Cell Biol. 2018, 102, 138–150. [Google Scholar] [CrossRef]
- Crea, F.; Quagliata, L.; Michael, A.; Liu, H.H.; Frumento, P.; Azad, A.A.; Xue, H.; Pikor, L.; Watahiki, A.; Morant, R.; et al. Integrated analysis of the prostate cancer small-nucleolar transcriptome reveals SNORA55 as a driver of prostate cancer progression. Mol. Oncol. 2016, 10, 693–703. [Google Scholar] [CrossRef] [Green Version]
- Hu, T.; Lu, C.; Xia, Y.; Wu, L.; Song, J.; Chen, C.; Wang, Q. Small nucleolar RNA SNORA71A promotes epithelial-mesenchymal transition by maintaining ROCK2 mRNA stability in breast cancer. Mol. Oncol. 2022, 16, 1947–1965. [Google Scholar] [CrossRef]
- Su, X.; Feng, C.; Wang, S.; Shi, L.; Gu, Q.; Zhang, H.; Lan, X.; Zhao, Y.; Qiang, W.; Ji, M.; et al. The noncoding RNAs SNORD50A and SNORD50B-mediated TRIM21-GMPS interaction promotes the growth of p53 wild-type breast cancers by degrading p53. Cell Death Differ. 2021, 28, 2450–2464. [Google Scholar] [CrossRef]
- Sun, Y.; Chen, E.; Li, Y.; Ye, D.; Cai, Y.; Wang, Q.; Li, Q.; Zhang, X. H/ACA box small nucleolar RNA 7B acts as an oncogene and a potential prognostic biomarker in breast cancer. Cancer Cell Int. 2019, 19, 125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mannoor, K.; Shen, J.; Liao, J.; Liu, Z.; Jiang, F. Small nucleolar RNA signatures of lung tumor-initiating cells. Mol. Cancer 2014, 13, 104. [Google Scholar] [CrossRef] [PubMed]
- Zheng, D.; Zhang, J.; Ni, J.; Luo, J.; Wang, J.; Tang, L.; Zhang, L.; Wang, L.; Xu, J.; Su, B.; et al. Small nucleolar RNA 78 promotes the tumorigenesis in non-small cell lung cancer. J. Exp. Clin. Cancer Res. 2015, 34, 49. [Google Scholar] [CrossRef] [Green Version]
- Yu, H.; Tian, L.; Yang, L.; Liu, S.; Wang, S.; Gong, J. Knockdown of SNORA47 Inhibits the Tumorigenesis of NSCLC via Mediation of PI3K/Akt Signaling Pathway. Front. Oncol. 2021, 11, 620213. [Google Scholar] [CrossRef]
- Tang, G.; Zeng, Z.; Sun, W.; Li, S.; You, C.; Tang, F.; Peng, S.; Ma, S.; Luo, Y.; Xu, J.; et al. Small Nucleolar RNA 71A Promotes Lung Cancer Cell Proliferation, Migration and Invasion via MAPK/ERK Pathway. J. Cancer 2019, 10, 2261–2275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, S.; Li, Q.; Chen, X.; Bao, H.; Wu, W.; Shen, F.; Lu, B.; Jiang, R.; Zong, Z.; Zhao, Y. SNORA70E promotes the occurrence and development of ovarian cancer through pseudouridylation modification of RAP1B and alternative splicing of PARPBP. J. Cell. Mol. Med. 2022, 26, 5150–5164. [Google Scholar] [CrossRef]
- Zhu, W.; Niu, J.; He, M.; Zhang, L.; Lv, X.; Liu, F.; Jiang, L.; Zhang, J.; Yu, Z.; Zhao, L.; et al. SNORD89 promotes stemness phenotype of ovarian cancer cells by regulating Notch1-c-Myc pathway. J. Transl. Med. 2019, 17, 259. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Ma, R.; Gao, M.; Zhao, Y.; Lv, X.; Zhu, W.; Han, L.; Su, P.; Fan, Y.; Yan, Y.; et al. SNORA72 Activates the Notch1/c-Myc Pathway to Promote Stemness Transformation of Ovarian Cancer Cells. Front. Cell Dev. Biol. 2020, 8, 583087. [Google Scholar] [CrossRef]
- Bao, H.J.; Chen, X.; Liu, X.; Wu, W.; Li, Q.H.; Xian, J.Y.; Zhao, Y.; Chen, S. Box C/D snoRNA SNORD89 influences the occurrence and development of endometrial cancer through 2’-O-methylation modification of Bim. Cell Death Discov. 2022, 8, 309. [Google Scholar] [CrossRef]
- Patterson, D.G.; Roberts, J.T.; King, V.M.; Houserova, D.; Barnhill, E.C.; Crucello, A.; Polska, C.J.; Brantley, L.W.; Kaufman, G.C.; Nguyen, M.; et al. Human snoRNA-93 is processed into a microRNA-like RNA that promotes breast cancer cell invasion. NPJ Breast Cancer 2017, 3, 25. [Google Scholar] [CrossRef] [Green Version]
- Heitzer, E.; Haque, I.S.; Roberts, C.E.S.; Speicher, M.R. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat. Rev. Genet. 2019, 20, 71–88. [Google Scholar] [CrossRef]
- Pinzani, P.; D’Argenio, V.; Del Re, M.; Pellegrini, C.; Cucchiara, F.; Salvianti, F.; Galbiati, S. Updates on liquid biopsy: Current trends and future perspectives for clinical application in solid tumors. Clin. Chem. Lab. Med. 2021, 59, 1181–1200. [Google Scholar] [CrossRef] [PubMed]
- Eichelser, C.; Stückrath, I.; Müller, V.; Milde-Langosch, K.; Wikman, H.; Pantel, K.; Schwarzenbach, H. Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget 2014, 5, 9650–9663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodríguez, M.; Ajona, D.; Seijo, L.M.; Sanz, J.; Valencia, K.; Corral, J.; Mesa-Guzmán, M.; Pío, R.; Calvo, A.; Lozano, M.D.; et al. Molecular biomarkers in early stage lung cancer. Transl. Lung Cancer Res. 2021, 10, 1165–1185. [Google Scholar] [CrossRef] [PubMed]
- Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef]
- Hu, X.; Macdonald, D.M.; Huettner, P.C.; Feng, Z.; El Naqa, I.M.; Schwarz, J.K.; Mutch, D.G.; Grigsby, P.W.; Powell, S.N.; Wang, X. A miR-200 microRNA cluster as prognostic marker in advanced ovarian cancer. Gynecol. Oncol. 2009, 114, 457–464. [Google Scholar] [CrossRef]
- Tan, C.; Cao, J.; Chen, L.; Xi, X.; Wang, S.; Zhu, Y.; Yang, L.; Ma, L.; Wang, D.; Yin, J.; et al. Noncoding RNAs Serve as Diagnosis and Prognosis Biomarkers for Hepatocellular Carcinoma. Clin. Chem. 2019, 65, 905–915. [Google Scholar] [CrossRef]
- Liao, J.; Yu, L.; Mei, Y.; Guarnera, M.; Shen, J.; Li, R.; Liu, Z.; Jiang, F. Small nucleolar RNA signatures as biomarkers for non-small-cell lung cancer. Mol. Cancer 2010, 9, 198. [Google Scholar] [CrossRef] [Green Version]
- Martens-Uzunova, E.S.; Hoogstrate, Y.; Kalsbeek, A.; Pigmans, B.; Vredenbregt-van den Berg, M.; Dits, N.; Nielsen, S.J.; Baker, A.; Visakorpi, T.; Bangma, C.; et al. C/D-box snoRNA-derived RNA production is associated with malignant transformation and metastatic progression in prostate cancer. Oncotarget 2015, 6, 17430–17444. [Google Scholar] [CrossRef] [Green Version]
- Rimer, J.M.; Lee, J.; Holley, C.; Crowder, R.J.; Chen, D.L.; Hanson, P.I.; Ory, D.S.; Schaffer, J.E. Long-range function of secreted small nucleolar RNAs that direct 2′-O-methylation. J. Biol. Chem. 2018, 293, 13284–13296. [Google Scholar] [CrossRef] [Green Version]
- Weber, J.A.; Baxter, D.H.; Zhang, S.; Huang, D.Y.; Huang, K.H.; Lee, M.J.; Galas, D.J.; Wang, K. The MicroRNA Spectrum in 12 Body Fluids. Clin. Chem. 2010, 56, 1733–1741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Umu, S.U.; Langseth, H.; Bucher-Johannessen, C.; Fromm, B.; Keller, A.; Meese, E.; Lauritzen, M.; Leithaug, M.; Lyle, R.; Rounge, T.B. A comprehensive profile of circulating RNAs in human serum. RNA Biol. 2018, 15, 242–250. [Google Scholar] [CrossRef] [PubMed]
- Balzano, F.; Deiana, M.; Giudici, S.D.; Oggiano, A.; Baralla, A.; Pasella, S.; Mannu, A.; Pescatori, M.; Porcu, B.; Fanciulli, G.; et al. miRNA Stability in Frozen Plasma Samples. Molecules 2015, 20, 19030–19040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mitchell, A.J.; Gray, W.D.; Hayek, S.S.; Ko, Y.-A.; Thomas, S.; Rooney, K.; Awad, M.; Roback, J.D.; Quyyumi, A.; Searles, C.D. Platelets confound the measurement of extracellular miRNA in archived plasma. Sci. Rep. 2016, 6, 32651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duttagupta, R.; Jiang, R.; Gollub, J.; Getts, R.C.; Jones, K.W. Impact of Cellular miRNAs on Circulating miRNA Biomarker Signatures. PLoS ONE 2011, 6, e20769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valones, M.A.A.; Guimarães, R.L.; Brandão, L.A.C.; de Souza, P.R.E.; Carvalho, A.D.A.T.; Crovela, S. Principles and applications of polymerase chain reaction in medical diagnostic fields: A review. Braz. J. Microbiol. 2009, 40, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Dodd, D.W.; Gagnon, K.T.; Corey, D.R. Digital Quantitation of Potential Therapeutic Target RNAs. Nucleic Acid Ther. 2013, 23, 188–194. [Google Scholar] [CrossRef] [Green Version]
- Link, J.C.; Hasin-Brumshtein, Y.; Cantor, R.M.; Chen, X.; Arnold, A.P.; Lusis, A.J.; Reue, K. Diet, gonadal sex, and sex chromosome complement influence white adipose tissue miRNA expression. BMC Genom. 2017, 18, 89. [Google Scholar] [CrossRef] [Green Version]
- Rounge, T.B.; Umu, S.U.; Keller, A.; Meese, E.; Ursin, G.; Tretli, S.; Lyle, R.; Langseth, H. Circulating small non-coding RNAs associated with age, sex, smoking, body mass and physical activity. Sci. Rep. 2018, 8, 17650. [Google Scholar] [CrossRef] [Green Version]
- Cheng, H.-Y.M.; Papp, J.W.; Varlamova, O.; Dziema, H.; Russell, B.; Curfman, J.P.; Nakazawa, T.; Shimizu, K.; Okamura, H.; Impey, S.; et al. microRNA Modulation of Circadian-Clock Period and Entrainment. Neuron 2007, 54, 813–829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turchinovich, A.; Drapkina, O.; Tonevitsky, A. Transcriptome of Extracellular Vesicles: State-of-the-Art. Front. Immunol. 2019, 10, 202. [Google Scholar] [CrossRef]
- Su, J.; Liao, J.; Gao, L.; Shen, J.; Guarnera, M.A.; Zhan, M.; Fang, H.; Stass, S.A.; Jiang, F. Analysis of small nucleolar RNAs in sputum for lung cancer diagnosis. Oncotarget 2016, 7, 5131–5142. [Google Scholar] [CrossRef] [Green Version]
- Shah, M.Y.; Ferrajoli, A.; Sood, A.K.; Lopez-Berestein, G.; Calin, G.A. microRNA Therapeutics in Cancer—An Emerging Concept. Ebiomedicine 2016, 12, 34–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gomarasca, M.; Maroni, P.; Banfi, G.; Lombardi, G. microRNAs in the Antitumor Immune Response and in Bone Metastasis of Breast Cancer: From Biological Mechanisms to Therapeutics. Int. J. Mol. Sci. 2020, 21, 2805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maroni, P.; Bendinelli, P.; Matteucci, E.; Desiderio, M.A. The therapeutic effect of miR-125b is enhanced by the prostaglandin endoperoxide synthase 2/cyclooxygenase 2 blockade and hampers ETS1 in the context of the microenvironment of bone metastasis. Cell Death Dis. 2018, 9, 472. [Google Scholar] [CrossRef] [Green Version]
- Momen-Heravi, F.; Bala, S.; Bukong, T.; Szabo, G. Exosome-mediated delivery of functionally active miRNA-155 inhibitor to macrophages. Nanomed. Nanotechnol. Biol. Med. 2014, 10, 1517–1527. [Google Scholar] [CrossRef] [Green Version]
- Hong, D.S.; Kang, Y.K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.L.; Kim, T.Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef]
- Reid, G.; Pel, M.; Kirschner, M.; Cheng, Y.; Mugridge, N.; Weiss, J.; Williams, M.; Wright, C.; Edelman, J.; Vallely, M.; et al. Restoring expression of miR-16: A novel approach to therapy for malignant pleural mesothelioma. Ann. Oncol. 2013, 24, 3128–3135. [Google Scholar] [CrossRef]
- Liang, X.-H.; Vickers, T.A.; Guo, S.; Crooke, S.T. Efficient and specific knockdown of small non-coding RNAs in mammalian cells and in mice. Nucleic Acids Res. 2011, 39, e13. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Puppo, M.; Jaafar, M.; Diaz, J.-J.; Marcel, V.; Clézardin, P. MiRNAs and snoRNAs in Bone Metastasis: Functional Roles and Clinical Potential. Cancers 2023, 15, 242. https://doi.org/10.3390/cancers15010242
Puppo M, Jaafar M, Diaz J-J, Marcel V, Clézardin P. MiRNAs and snoRNAs in Bone Metastasis: Functional Roles and Clinical Potential. Cancers. 2023; 15(1):242. https://doi.org/10.3390/cancers15010242
Chicago/Turabian StylePuppo, Margherita, Mariam Jaafar, Jean-Jacques Diaz, Virginie Marcel, and Philippe Clézardin. 2023. "MiRNAs and snoRNAs in Bone Metastasis: Functional Roles and Clinical Potential" Cancers 15, no. 1: 242. https://doi.org/10.3390/cancers15010242
APA StylePuppo, M., Jaafar, M., Diaz, J. -J., Marcel, V., & Clézardin, P. (2023). MiRNAs and snoRNAs in Bone Metastasis: Functional Roles and Clinical Potential. Cancers, 15(1), 242. https://doi.org/10.3390/cancers15010242