Identification of Exo-miRNAs: A Summary of the Efforts in Translational Studies Involving Triple-Negative Breast Cancer
Abstract
:1. Introduction
2. Materials and Methods
3. Exo-miR Biology: Formation and Role in TNBC
3.1. Exosome Isolation and Evaluation of Exosomal Markers
3.2. Special Considerations: Non-Exosomal Contamination and Data Normalization
4. Benefits of Including a Discovery Cohort with a Control Group in Translational Studies
4.1. Including a Control Group
4.2. Discovery Experiment
5. Current Efforts on the Development of a Liquid Biopsy Tool for TNBC Detection
Study | Discovery Experiment | Validation Cohort | Isolation Method | Detection Method | Study Results |
---|---|---|---|---|---|
Wu et al. (2020) [76] | Yes (n = 30) | Yes (n = 40) | Exosome Isolation Reagent | Next generation sequencing and RT-qPCR | Upregulated miRNAs were miR-148a-5p, miR-200a-5p, miR-210a-3p, miR-378a-3p, miR-483-5p and miR-7110-5p). Downregulated miRNAs were: miR-92b-3p and miR-150-5p. |
Stevic et al. (2018) [75] | Yes (n =15) | Yes (n = 455) | ExoQuick Exosome Precipitation Solution | TaqMan microRNA array Human Pool A cards and TaqMan RT-PCR | Significant differences on 31 of the targets were detected among BC subtypes (HER2+ and TNBC). A significant association was found between exo-miR expression levels and tumor characteristics. |
Ni et al. (2018) [99] | Yes (n = 48) | Yes (n = 192) | ExoQuick Exosome Precipitation Solution | TaqMan miRNA array cards and TaqMan miRNA assays | miR-16, miR-30b, and miR-93 have specific exosome packaging. Levels of miR-93 were significantly enriched in exosomes from DCIS patients rather than BC patients. Levels of miR-16 were high in patients with ER+ (n = 85) when compared to TNBC patients (n = 24). |
Li et al. (2018) [102] | Yes Plasma (n = 400) Serum (n = 406) samples | Yes (n = 32) Exosome extraction | ExoQuick Exosome Precipitation Solution | qRT-PCR | miR-20b-5p was significantly upregulated in BC. Exo-miR-106a-5p was consistently reported across compartments. No potential results related to TNBC subtype were reported, although BC subtypes were considered in the experimental design. |
Eichelser et al. (2014) [106] | Yes (n = 215) | Yes (n = 62) | ExoQuick Exosome Precipitation Solution | TaqMan MicroRNA Assays | miR-101, miR-372, and miR-373 were found in higher expression in the exo-miR fraction when compared with cell-free miRNAs. miR-373 was highly expressed on TNBC samples |
Hannafon et al. (2016) [77] | Combination of in vitro and PDX mice | Yes (n = 32) | ExoQuick Exosome Precipitation Solution | qRT-PCR | miR-1246 and miR-21 were significantly highly expressed on BC patients. ROC: miR-1246 (0.69), miR-21 (0.69), and the combination (0.73). |
Yoshikawa et al. (2018) [107] | Yes (n = 9) | Yes (n = 199) | Ultracentrifugation | TaqMan RT-qPCR | exo-miR-223-3p was higher in BC cases and controls. Exo-miR-223-3p expression was associated with the histological type, pT stage, pN stage, pathological stage, lymphatic invasion, and nuclear grade. |
6. The Use of In Vitro Models to Study Exo-miR Mediated Mechanisms
7. Future Perspectives to Advance TNBC and Liquid Biopsy
8. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Almansour, N.M. Triple-Negative Breast Cancer: A Brief Review About Epidemiology, Risk Factors, Signaling Pathways, Treatment and Role of Artificial Intelligence. Front. Mol. Biosci. 2022, 9, 836417. [Google Scholar] [CrossRef] [PubMed]
- Aysola, K.; Desai, A.; Welch, C.; Xu, J.; Qin, Y.; Reddy, V.; Matthews, R.; Owens, C.; Okoli, J.; Beech, D.; et al. Triple Negative Breast Cancer—An Overview. Hered. Genet. 2013, 2013 (Suppl. S2), 001. [Google Scholar]
- Jitariu, A.-A.; Cîmpean, A.M.; Ribatti, D.; Raica, M. Triple negative breast cancer: The kiss of death. Oncotarget 2017, 8, 46652–46662. [Google Scholar] [CrossRef] [PubMed]
- Elsawaf, Z.; Sinn, H.P. Triple-Negative Breast Cancer: Clinical and Histological Correlations. Breast Care 2011, 6, 273–278. [Google Scholar] [CrossRef] [PubMed]
- Al-Mahmood, S.; Sapiezynski, J.; Garbuzenko, O.B.; Minko, T. Metastatic and triple-negative breast cancer: Challenges and treatment options. Drug Deliv. Transl. Res. 2018, 8, 1483–1507. [Google Scholar] [CrossRef]
- Yin, L.; Duan, J.-J.; Bian, X.-W.; Yu, S.-C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020, 22, 61. [Google Scholar] [CrossRef]
- Ensenyat-Mendez, M.; Llinàs-Arias, P.; Orozco, J.I.J.; Íñiguez-Muñoz, S.; Salomon, M.P.; Sesé, B.; DiNome, M.L.; Marzese, D.M. Current Triple-Negative Breast Cancer Subtypes: Dissecting the Most Aggressive Form of Breast Cancer. Front. Oncol. 2021, 11, 681476. [Google Scholar] [CrossRef]
- Bou Zerdan, M.; Ghorayeb, T.; Saliba, F.; Allam, S.; Yaghi, M.; Bilani, N.; Jaafar, R.; Nahleh, Z. Triple Negative Breast Cancer: Updates on Classification and Treatment in 2021. Cancers 2022, 14, 1253. [Google Scholar] [CrossRef]
- Hubbell, E.; Clarke, C.A.; Aravanis, A.M.; Berg, C.D. Modeled Reductions in Late-stage Cancer with a Multi-Cancer Early Detection Test. Cancer Epidemiol. Biomark. Prev. 2021, 30, 460–468. [Google Scholar] [CrossRef]
- Summary Report of the Working Group to Review the National Cancer Institute-American Cancer Society Breast Cancer Detection Demonstration Projects. JNCI J. Natl. Cancer Inst. 1979, 62, 647–650. [CrossRef]
- Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA A Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
- Halvaei, S.; Daryani, S.; Eslami-S, Z.; Samadi, T.; Jafarbeik-Iravani, N.; Bakhshayesh, T.O.; Majidzadeh-A, K.; Esmaeili, R. Exosomes in Cancer Liquid Biopsy: A Focus on Breast Cancer. Mol. Ther.—Nucleic Acids 2017, 10, 131–141. [Google Scholar] [CrossRef]
- Zhou, B.; Xu, K.; Zheng, X.; Chen, T.; Wang, J.; Song, Y.; Shao, Y.; Zheng, S. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct. Target. Ther. 2020, 5, 144. [Google Scholar] [CrossRef] [PubMed]
- Shao, H.; Chung, J.; Issadore, D. Diagnostic technologies for circulating tumour cells and exosomes. Biosci. Rep. 2016, 36, e00292. [Google Scholar] [CrossRef] [PubMed]
- Annese, T.; Tamma, R.; De Giorgis, M.; Ribatti, D. microRNAs Biogenesis, Functions and Role in Tumor Angiogenesis. Front. Oncol. 2020, 10, 581007. [Google Scholar] [CrossRef]
- Lin, S.; Gregory, R.I. MicroRNA biogenesis pathways in cancer. Nat. Rev. Cancer 2015, 15, 321–333. [Google Scholar] [CrossRef] [PubMed]
- O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef]
- Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef]
- Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
- Baixauli, F.; Lopez-Otin, C.; Mittelbrunn, M. Exosomes and autophagy: Coordinated mechanisms for the maintenance of cellular fitness. Front. Immunol. 2014, 5, 403. [Google Scholar] [CrossRef]
- Kakarla, R.; Hur, J.; Kim, Y.J.; Kim, J.; Chwae, Y.-J. Apoptotic cell-derived exosomes: Messages from dying cells. Exp. Mol. Med. 2020, 52, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Delpech, J.-C.; Herron, S.; Botros, M.B.; Ikezu, T. Neuroimmune Crosstalk through Extracellular Vesicles in Health and Disease. Trends Neurosci. 2019, 42, 361–372. [Google Scholar] [CrossRef] [PubMed]
- van Balkom, B.W.; De Jong, O.G.; Smits, M.; Brummelman, J.; den Ouden, K.; de Bree, P.M.; van Eijndhoven, M.A.J.; Pegtel, D.M.; Stoorvogel, W.; Würdinger, T.; et al. Endothelial cells require miR-214 to secrete exosomes that suppress senescence and induce angiogenesis in human and mouse endothelial cells. Blood 2013, 121, 3997–4006. [Google Scholar] [CrossRef] [PubMed]
- Hussain, M.W.A.; Jahangir, S.; Ghosh, B.; Yesmin, F.; Anis, A.; Satil, S.N.; Anwar, F.; Rashid, M.H. Exosomes for Regulation of Immune Responses and Immunotherapy. J. Nanotheranostics 2022, 3, 55–85. [Google Scholar] [CrossRef]
- Schwarzenbach, H.; Gahan, P.B. Exosomes in Immune Regulation. Non-Coding RNA 2021, 7, 4. [Google Scholar] [CrossRef] [PubMed]
- Paskeh, M.D.A.; Entezari, M.; Mirzaei, S.; Zabolian, A.; Saleki, H.; Naghdi, M.J.; Sabet, S.; Khoshbakht, M.A.; Hashemi, M.; Hushmandi, K.; et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J. Hematol. Oncol. 2022, 15, 83. [Google Scholar] [CrossRef]
- Ge, L.; Zhou, F.; Nie, J.; Wang, X.; Zhao, Q. Hypoxic colorectal cancer-secreted exosomes deliver miR-210-3p to normoxic tumor cells to elicit a protumoral effect. Exp. Biol. Med. 2021, 246, 1895–1906. [Google Scholar] [CrossRef]
- 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]
- Liu, Q.; Peng, F.; Chen, J. The Role of Exosomal MicroRNAs in the Tumor Microenvironment of Breast Cancer. Int. J. Mol. Sci. 2019, 20, 3884. [Google Scholar] [CrossRef] [PubMed]
- Felekkis, K.; Touvana, E.; Stefanou, C.; Deltas, C. microRNAs: A newly described class of encoded molecules that play a role in health and disease. Hippokratia 2010, 14, 236–240. [Google Scholar]
- Colombo, M.; Moita, C.; van Niel, G.; Kowal, J.; Vigneron, J.; Benaroch, P.; Manel, N.; Moita, L.F.; Théry, C.; Raposo, G. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 2013, 126, 5553–5565. [Google Scholar] [CrossRef]
- Andreu, Z.; Yanez-Mo, M. Tetraspanins in extracellular vesicle formation and function. Front. Immunol. 2014, 5, 442. [Google Scholar] [CrossRef]
- Dedes, K.J.; Natrajan, R.; Lambros, M.B.; Geyer, F.C.; Lopez-Garcia, M.A.; Savage, K.; Jones, R.L.; Reis-Filho, J.S. Down-regulation of the miRNA master regulators Drosha and Dicer is associated with specific subgroups of breast cancer. Eur. J. Cancer 2011, 47, 138–150. [Google Scholar] [CrossRef]
- Passon, N.; Gerometta, A.; Puppin, C.; Lavarone, E.; Puglisi, F.; Tell, G.; Di Loreto, C.; Damante, G. Expression of Dicer and Drosha in triple-negative breast cancer. J. Clin. Pathol. 2012, 65, 320–326. [Google Scholar] [CrossRef] [PubMed]
- Dilsiz, N. Role of exosomes and exosomal microRNAs in cancer. Futur. Sci. OA 2020, 6, FSO465. [Google Scholar] [CrossRef] [PubMed]
- Santos, J.C.; da Silva Lima, N.; Sarian, L.O.; Matheu, A.; Ribeiro, M.L.; Derchain, S.F.M. Exosome-mediated breast cancer chemoresistance via miR-155 transfer. Sci. Rep. 2018, 8, 829. [Google Scholar] [CrossRef]
- Yang, S.-S.; Ma, S.; Dou, H.; Liu, F.; Zhang, S.-Y.; Jiang, C.; Xiao, M.; Huang, Y.-X. Breast cancer-derived exosomes regulate cell invasion and metastasis in breast cancer via miR-146a to activate cancer associated fibroblasts in tumor microenvironment. Exp. Cell Res. 2020, 391, 111983. [Google Scholar] [CrossRef] [PubMed]
- Doyle, L.; Wang, M. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed]
- Larios, J.; Mercier, V.; Roux, A.; Gruenberg, J. ALIX- and ESCRT-III–dependent sorting of tetraspanins to exosomes. J. Cell Biol. 2020, 219, 3. [Google Scholar] [CrossRef]
- Gastpar, R.; Gehrmann, M.; Bausero, M.A.; Asea, A.; Gross, C.; Schroeder, J.A.; Multhoff, G. Heat Shock Protein 70 Surface-Positive Tumor Exosomes Stimulate Migratory and Cytolytic Activity of Natural Killer Cells. Cancer Res 2005, 65, 5238–5247. [Google Scholar] [CrossRef]
- Crenshaw, B.J.; Kumar, S.; Bell, C.R.; Jones, L.B.; Williams, S.D.; Saldanha, S.N.; Joshi, S.; Sahu, R.; Sims, B.; Matthews, Q.L. Alcohol Modulates the Biogenesis and Composition of Microglia-Derived Exosomes. Biology 2019, 8, 25. [Google Scholar] [CrossRef]
- Sun, S.; Sun, L.; Zhou, X.; Wu, C.; Wang, R.; Lin, S.-H.; Kuang, J. Phosphorylation-Dependent Activation of the ESCRT Function of ALIX in Cytokinetic Abscission and Retroviral Budding. Dev. Cell 2016, 36, 331–343. [Google Scholar] [CrossRef]
- Sahu, R.; Kaushik, S.; Clement, C.C.; Cannizzo, E.S.; Scharf, B.; Follenzi, A.; Potolicchio, I.; Nieves, E.; Cuervo, A.M.; Santambrogio, L. Microautophagy of cytosolic proteins by late endosomes. Dev. Cell 2011, 20, 131–139. [Google Scholar] [CrossRef]
- Yoshimura, C.; Nagatoishi, S.; Kuroda, D.; Kodama, Y.; Uno, T.; Kitade, M.; Chong-Takata, K.; Oshiumi, H.; Muraoka, H.; Yamashita, S.; et al. Thermodynamic Dissection of Potency and Selectivity of Cytosolic Hsp90 Inhibitors. J. Med. Chem. 2021, 64, 2669–2677. [Google Scholar] [CrossRef]
- Escola, J.-M.; Kleijmeer, M.J.; Stoorvogel, W.; Griffith, J.M.; Yoshie, O.; Geuze, H.J. Selective Enrichment of Tetraspan Proteins on the Internal Vesicles of Multivesicular Endosomes and on Exosomes Secreted by Human B-lymphocytes. J. Biol. Chem. 1998, 273, 20121–20127. [Google Scholar] [CrossRef]
- Semina, S.E.; Scherbakov, A.M.; Vnukova, A.A.; Bagrov, D.V.; Evtushenko, E.G.; Safronova, V.M.; Golovina, D.A.; Lyubchenko, L.N.; Gudkova, M.V.; Krasil’Nikov, M.A. Exosome-Mediated Transfer of Cancer Cell Resistance to Antiestrogen Drugs. Molecules 2018, 23, 829. [Google Scholar] [CrossRef] [PubMed]
- Muralidharan-Chari, V.; Clancy, J.; Plou, C.; Romao, M.; Chavrier, P.; Raposo, G.; D’Souza-Schorey, C. ARF6-Regulated Shedding of Tumor Cell-Derived Plasma Membrane Microvesicles. Curr. Biol. 2009, 19, 1875–1885. [Google Scholar] [CrossRef] [PubMed]
- Akers, J.C.; Gonda, D.; Kim, R.; Carter, B.S.; Chen, C.C. Biogenesis of extracellular vesicles (EV): Exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J. Neuro-Oncol. 2013, 113, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Patel, G.K.; Khan, M.A.; Zubair, H.; Srivastava, S.K.; Khushman, M.; Singh, S.; Singh, A. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Sci. Rep. 2019, 9, 5335. [Google Scholar] [CrossRef] [PubMed]
- Moon, S.; Shin, D.W.; Kim, S.; Lee, Y.-S.; Mankhong, S.; Yang, S.W.; Lee, P.H.; Park, D.-H.; Kwak, H.-B.; Lee, J.-S.; et al. Enrichment of Exosome-Like Extracellular Vesicles from Plasma Suitable for Clinical Vesicular miRNA Biomarker Research. J. Clin. Med. 2019, 8, 1995. [Google Scholar] [CrossRef]
- Sung, B.H.; von Lersner, A.; Guerrero, J.; Krystofiak, E.S.; Inman, D.; Pelletier, R.; Zijlstra, A.; Ponik, S.M.; Weaver, A.M. A live cell reporter of exosome secretion and uptake reveals pathfinding behavior of migrating cells. Nat. Commun. 2020, 11, 2092. [Google Scholar] [CrossRef]
- Rupp, A.-K.; Rupp, C.; Keller, S.; Brase, J.C.; Ehehalt, R.; Fogel, M.; Moldenhauer, G.; Marmé, F.; Sültmann, H.; Altevogt, P. Loss of EpCAM expression in breast cancer derived serum exosomes: Role of proteolytic cleavage. Gynecol. Oncol. 2011, 122, 437–446. [Google Scholar] [CrossRef] [PubMed]
- Khan, S.; Bennit, H.F.; Turay, D.; Perez, M.; Mirshahidi, S.; Yuan, Y.; Wall, N.R. Early diagnostic value of survivin and its alternative splice variants in breast cancer. BMC Cancer 2014, 14, 176. [Google Scholar] [CrossRef] [PubMed]
- Ciravolo, V.; Huber, V.; Ghedini, G.C.; Venturelli, E.; Bianchi, F.; Campiglio, M.; Morelli, D.; Villa, A.; Della Mina, P.; Menard, S.; et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J. Cell. Physiol. 2011, 227, 658–667. [Google Scholar] [CrossRef]
- Fang, S.; Tian, H.; Li, X.; Jin, D.; Li, X.; Kong, J.; Yang, C.; Yang, X.; Lu, Y.; Luo, Y.; et al. Clinical application of a microfluidic chip for immunocapture and quantification of circulating exosomes to assist breast cancer diagnosis and molecular classification. PLoS ONE 2017, 12, e0175050. [Google Scholar] [CrossRef]
- Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gammon, S.T.; Kaye, J.; LeBleu, V.S.; Mittendorf, E.A.; Weitz, J.; Rahbari, N.; et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 2015, 523, 177–182. [Google Scholar] [CrossRef]
- Vardaki, I.; Ceder, S.; Rutishauser, D.; Baltatzis, G.; Foukakis, T.; Panaretakis, T. Periostin is identified as a putative metastatic marker in breast cancer-derived exosomes. Oncotarget 2016, 7, 74966–74978. [Google Scholar] [CrossRef] [PubMed]
- Kibria, G.; Ramos, E.K.; Lee, K.E.; Bedoyan, S.; Huang, S.; Samaeekia, R.; Athman, J.J.; Harding, C.V.; Lötvall, J.; Harris, L.; et al. A rapid, automated surface protein profiling of single circulating exosomes in human blood. Sci. Rep. 2016, 6, 36502. [Google Scholar] [CrossRef]
- Gandham, S.; Su, X.; Wood, J.; Nocera, A.L.; Alli, S.C.; Milane, L.; Zimmerman, A.; Amiji, M.; Ivanov, A.R. Technologies and Standardization in Research on Extracellular Vesicles. Trends Biotechnol. 2020, 38, 1066–1098. [Google Scholar] [CrossRef] [PubMed]
- Tellez-Gabriel, M.; Knutsen, E.; Perander, M. Current Status of Circulating Tumor Cells, Circulating Tumor DNA, and Exosomes in Breast Cancer Liquid Biopsies. Int. J. Mol. Sci. 2020, 21, 9457. [Google Scholar] [CrossRef] [PubMed]
- Ludwig, N.; Whiteside, T.L.; Reichert, T.E. Challenges in Exosome Isolation and Analysis in Health and Disease. Int. J. Mol. Sci. 2019, 20, 4684. [Google Scholar] [CrossRef] [PubMed]
- Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef]
- Kurian, T.K.; Banik, S.; Gopal, D.; Chakrabarti, S.; Mazumder, N. Elucidating Methods for Isolation and Quantification of Exosomes: A Review. Mol. Biotechnol. 2021, 63, 249–266. [Google Scholar] [CrossRef] [PubMed]
- Danielson, K.; Estanislau, J.; Tigges, J.; Toxavidis, V.; Camacho, V.; Felton, E.J.; Khoory, J.; Kreimer, S.; Ivanov, A.R.; Mantel, P.-Y.; et al. Diurnal Variations of Circulating Extracellular Vesicles Measured by Nano Flow Cytometry. PLoS ONE 2016, 11, e0144678. [Google Scholar] [CrossRef] [PubMed]
- Xu, G.; Zhang, B.; Ye, J.; Cao, S.; Shi, J.; Zhao, Y.; Wang, Y.; Sang, J.; Yao, Y.; Guan, W.; et al. Exosomal miRNA-139 in cancer-associated fibroblasts inhibits gastric cancer progression by repressing MMP11 expression. Int. J. Biol. Sci. 2019, 15, 2320–2329. [Google Scholar] [CrossRef]
- He, L.; Zhu, W.; Chen, Q.; Yuan, Y.; Wang, Y.; Wang, J.; Wu, X. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics 2019, 9, 8206–8220. [Google Scholar] [CrossRef]
- Damanti, C.C.; Gaffo, E.; Lovisa, F.; Garbin, A.; Di Battista, P.; Gallingani, I.; Tosato, A.; Pillon, M.; Carraro, E.; Mascarin, M.; et al. MiR-26a-5p as a Reference to Normalize MicroRNA qRT-PCR Levels in Plasma Exosomes of Pediatric Hematological Malignancies. Cells 2021, 10, 101. [Google Scholar] [CrossRef]
- Faraldi, M.; Gomarasca, M.; Sansoni, V.; Perego, S.; Banfi, G.; Lombardi, G. Normalization strategies differently affect circulating miRNA profile associated with the training status. Sci. Rep. 2019, 9, 1584. [Google Scholar] [CrossRef] [PubMed]
- Sanz-Rubio, D.; Martin-Burriel, I.; Gil, A.; Cubero, P.; Forner, M.; Khalyfa, A.; Marin, J.M. Stability of Circulating Exosomal miRNAs in Healthy Subjects. Sci. Rep. 2018, 8, 10306. [Google Scholar] [CrossRef]
- Gheinani, A.H.; Vögeli, M.; Baumgartner, U.; Vassella, E.; Draeger, A.; Burkhard, F.C.; Monastyrskaya, K. Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery. Sci. Rep. 2018, 8, 3945. [Google Scholar] [CrossRef]
- Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef] [PubMed]
- Grigor’eva, A.E.; Tamkovich, S.N.; Eremina, A.V.; Tupikin, A.E.; Kabilov, M.R.; Chernykh, V.V.; Vlassov, V.V.; Laktionov, P.P.; Ryabchikova, E.I. Exosomes in tears of healthy individuals: Isolation, identification, and characterization. Biochem. Suppl. Ser. B Biomed. Chem. 2016, 10, 165–172. [Google Scholar] [CrossRef]
- Khan, S.; Simpson, J.; Lynch, J.C.; Turay, D.; Mirshahidi, S.; Gonda, A.; Sanchez, T.W.; Casiano, C.A.; Wall, N.R. Racial differences in the expression of inhibitors of apoptosis (IAP) proteins in extracellular vesicles (EV) from prostate cancer patients. PLoS ONE 2017, 12, e0183122. [Google Scholar] [CrossRef]
- Hooten, N.N.; McFarland, M.H.; Freeman, D.W.; Mode, N.A.; Ezike, N.; Zonderman, A.B.; Evans, M.K. Association of Extracellular Vesicle Protein Cargo with Race and Clinical Markers of Mortality. Sci. Rep. 2019, 9, 17582. [Google Scholar] [CrossRef]
- Stevic, I.; Müller, V.; Weber, K.; Fasching, P.A.; Karn, T.; Marmé, F.; Schem, C.; Stickeler, E.; Denkert, C.; Van Mackelenbergh, M.; et al. Specific microRNA signatures in exosomes of triple-negative and HER2-positive breast cancer patients undergoing neoadjuvant therapy within the GeparSixto trial. BMC Med. 2018, 16, 179. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Wang, Q.; Zhong, H.; Li, L.; Zhang, Q.; Huang, Q.; Yu, Z. Differentially expressed microRNAs in exosomes of patients with breast cancer revealed by next-generation sequencing. Oncol. Rep. 2019, 43, 240–250. [Google Scholar] [CrossRef]
- Hannafon, B.N.; Trigoso, Y.D.; Calloway, C.L.; Zhao, Y.D.; Lum, D.H.; Welm, A.L.; Zhao, Z.J.; Blick, K.E.; Dooley, W.C.; Ding, W.Q. Plasma exosome microRNAs are indicative of breast cancer. Breast Cancer Res. 2016, 18, 90. [Google Scholar] [CrossRef]
- Pepe, M.S.; Feng, Z. Improving Biomarker Identification with Better Designs and Reporting. Clin. Chem. 2011, 57, 1093–1095. [Google Scholar] [CrossRef]
- Estevão-Pereira, H.; Lobo, J.; Salta, S.; Amorim, M.; Lopes, P.; Cantante, M.; Reis, B.; Antunes, L.; Castro, F.; De Sousa, S.P.; et al. Overexpression of circulating MiR-30b-5p identifies advanced breast cancer. J. Transl. Med. 2019, 17, 435. [Google Scholar] [CrossRef]
- Adam-Artigues, A.; Garrido-Cano, I.; Simón, S.; Ortega, B.; Moragón, S.; Lameirinhas, A.; Constâncio, V.; Salta, S.; Burgués, O.; Bermejo, B.; et al. Circulating miR-30b-5p levels in plasma as a novel potential biomarker for early detection of breast cancer. ESMO Open 2021, 6, 100039. [Google Scholar] [CrossRef]
- Dobbin, K.K.; Simon, R.M. Sample size planning for developing classifiers using high-dimensional DNA microarray data. Biostatistics 2006, 8, 101–117. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.; Grennan, K.; Badner, J.; Zhang, D.; Gershon, E.; Jin, L.; Liu, C. Removing Batch Effects in Analysis of Expression Microarray Data: An Evaluation of Six Batch Adjustment Methods. PLoS ONE 2011, 6, e17238. [Google Scholar] [CrossRef]
- Nayak, B.K. Understanding the relevance of sample size calculation. Indian J. Ophthalmol. 2010, 58, 469–470. [Google Scholar] [CrossRef]
- Agarwal, V.; Bell, G.W.; Nam, J.W.; Bartel, D.P. Predicting effective microRNA target sites in mammalian mRNAs. Elife 2015, 4, e05005. [Google Scholar] [CrossRef]
- Reczko, M.; Maragkakis, M.; Alexiou, P.; Grosse, I.; Hatzigeorgiou, A.G. Functional microRNA targets in protein coding sequences. Bioinformatics 2012, 28, 771–776. [Google Scholar] [CrossRef]
- Vlachos, I.S.; Zagganas, K.; Paraskevopoulou, M.D.; Georgakilas, G.; Karagkouni, D.; Vergoulis, T.; Dalamagas, T.; Hatzigeorgiou, A.G. DIANA-miRPath v3.0: Deciphering microRNA function with experimental support. Nucleic Acids Res. 2015, 43, W460–W466. [Google Scholar] [CrossRef] [PubMed]
- Xiao, F.; Zuo, Z.; Cai, G.; Kang, S.; Gao, X.; Li, T. miRecords: An integrated resource for microRNA-target interactions. Nucleic Acids Res. 2009, 37 (Suppl. S1), D105–D110. [Google Scholar] [CrossRef] [PubMed]
- Cho, S.; Jang, I.; Jun, Y.; Yoon, S.; Ko, M.; Kwon, Y.; Choi, I.; Chang, H.; Ryu, D.; Lee, B.; et al. miRGator v3.0: A microRNA portal for deep sequencing, expression profiling and mRNA targeting. Nucleic Acids Res. 2012, 41, D252–D257. [Google Scholar] [CrossRef] [PubMed]
- Sticht, C.; Gretz, N. In-Silico Algorithms for the Screening of Possible microRNA Binding Sites and Their Interactions. Curr. Genom. 2013, 14, 127–136. [Google Scholar] [CrossRef]
- Liu, W.; Wang, X. Prediction of functional microRNA targets by integrative modeling of microRNA binding and target expression data. Genome Biol. 2019, 20, 18. [Google Scholar] [CrossRef]
- Riolo, G.; Cantara, S.; Marzocchi, C.; Ricci, C. miRNA Targets: From Prediction Tools to Experimental Validation. Methods Protoc. 2020, 4, 1. [Google Scholar] [CrossRef] [PubMed]
- Livshits, M.A.; Khomyakova, E.; Evtushenko, E.G.; Lazarev, V.N.; Kulemin, N.A.; Semina, S.E.; Generozov, E.V.; Govorun, V.M. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci. Rep. 2015, 5, 17319. [Google Scholar] [CrossRef] [PubMed]
- Tajadini, M.; Panjehpour, M.; Javanmard, S.H. Comparison of SYBR Green and TaqMan methods in quantitative real-time polymerase chain reaction analysis of four adenosine receptor subtypes. Adv. Biomed. Res. 2014, 3, 85. [Google Scholar] [CrossRef] [PubMed]
- Ogata, H.; Goto, S.; Sato, K.; Fujibuchi, W.; Bono, H.; Kanehisa, M. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 2000, 28, 29–34. [Google Scholar] [CrossRef] [PubMed]
- Encarnación-Medina, J.; Ortiz, C.; Vergne, R.; Padilla, L.; Matta, J. MicroRNA Expression Changes in Women with Breast Cancer Stratified by DNA Repair Capacity Levels. J. Oncol. 2019, 2019, 7820275. [Google Scholar] [CrossRef]
- Tang, W.; Xu, P.; Wang, H.; Niu, Z.; Zhu, D.; Lin, Q.; Tang, L.; Ren, L. MicroRNA-150 suppresses triple-negative breast cancer metastasis through targeting HMGA2. OncoTargets Ther. 2018, 11, 2319–2332. [Google Scholar] [CrossRef]
- von Minckwitz, G.; Schneeweiss, A.; Loibl, S.; Salat, C.; Denkert, C.; Rezai, M.; Blohmer, J.; Jackisch, C.; Paepke, S.; Gerber, B.; et al. Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer (GeparSixto; GBG 66): A randomised phase 2 trial. Lancet Oncol. 2014, 15, 747–756. [Google Scholar] [CrossRef]
- Loibl, S.; Weber, K.E.; Timms, K.M.; Elkin, E.P.; Hahnen, E.; Fasching, P.A.; Lederer, B.; Denkert, C.; Schneeweiss, A.; Braun, S.; et al. Survival analysis of carboplatin added to an anthracycline/taxane-based neoadjuvant chemotherapy and HRD score as predictor of response—Final results from GeparSixto. Ann. Oncol. 2018, 29, 2341–2347. [Google Scholar] [CrossRef]
- Ni, Q.; Stevic, I.; Pan, C.; Müller, V.; Oliveira-Ferrer, L.; Pantel, K.; Schwarzenbach, H. Different signatures of miR-16, miR-30b and miR-93 in exosomes from breast cancer and DCIS patients. Sci. Rep. 2018, 8, 12974. [Google Scholar] [CrossRef]
- Xiang, M.; Zeng, Y.; Yang, R.; Xu, H.; Chen, Z.; Zhong, J.; Xie, H.; Xu, Y.; Zeng, X. U6 is not a suitable endogenous control for the quantification of circulating microRNAs. Biochem. Biophys. Res. Commun. 2014, 454, 210–214. [Google Scholar] [CrossRef]
- Schwarzenbach, H.; Da Silva, A.M.; Calin, G.; Pantel, K. Data Normalization Strategies for MicroRNA Quantification. Clin. Chem. 2015, 61, 1333–1342. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Zhou, Y.; Xia, T.; Zhou, X.; Huang, Z.; Zhang, H.; Zhu, W.; Ding, Q.; Wang, S. Circulating microRNAs from the miR-106a–363 cluster on chromosome X as novel diagnostic biomarkers for breast cancer. Breast Cancer Res. Treat. 2018, 170, 257–270. [Google Scholar] [CrossRef] [PubMed]
- Eichelser, C.; Flesch-Janys, D.; Chang-Claude, J.; Pantel, K.; Schwarzenbach, H. Deregulated serum concentrations of circulating cell-free microRNAs miR-17, miR-34a, miR-155, and miR-373 in human breast cancer development and progression. Clin. Chem. 2013, 59, 1489–1496. [Google Scholar] [CrossRef] [PubMed]
- Sachdeva, M.; Wu, H.; Ru, P.; Hwang, L.; Trieu, V.; Mo, Y.-Y. MicroRNA-101-mediated Akt activation and estrogen-independent growth. Oncogene 2010, 30, 822–831. [Google Scholar] [CrossRef]
- Zhou, A.-D.; Diao, L.-T.; Xu, H.; Xiao, Z.-D.; Li, J.-H.; Zhou, H.; Qu, L.-H. β-Catenin/LEF1 transactivates the microRNA-371-373 cluster that modulates the Wnt/β-catenin-signaling pathway. Oncogene 2011, 31, 2968–2978. [Google Scholar] [CrossRef]
- 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]
- Yoshikawa, M.; Iinuma, H.; Umemoto, Y.; Yanagisawa, T.; Matsumoto, A.; Jinno, H. Exosome-encapsulated microRNA-223-3p as a minimally invasive biomarker for the early detection of invasive breast cancer. Oncol. Lett. 2018, 15, 9584–9592. [Google Scholar] [CrossRef]
- O’brien, K.; Rani, S.; Corcoran, C.; Wallace, R.; Hughes, L.; Friel, A.M.; McDonnell, S.; Crown, J.; Radomski, M.W.; O’driscoll, L. Exosomes from triple-negative breast cancer cells can transfer phenotypic traits representing their cells of origin to secondary cells. Eur. J. Cancer 2013, 49, 1845–1859. [Google Scholar] [CrossRef]
- Gernapudi, R.; Yao, Y.; Zhang, Y.; Wolfson, B.; Roy, S.; Duru, N.; Eades, G.; Yang, P.; Zhou, Q. Targeting exosomes from preadipocytes inhibits preadipocyte to cancer stem cell signaling in early-stage breast cancer. Breast Cancer Res. Treat. 2015, 150, 685–695. [Google Scholar] [CrossRef]
- Thiese, M.S. Observational and interventional study design types; an overview. Biochem. Medica 2014, 24, 199–210. [Google Scholar] [CrossRef]
- Dumas-Mallet, E.; Button, K.S.; Boraud, T.; Gonon, F.; Munafò, M.R. Low statistical power in biomedical science: A review of three human research domains. R. Soc. Open Sci. 2017, 4, 160254. [Google Scholar] [CrossRef]
- Burd, E.M. Validation of Laboratory-Developed Molecular Assays for Infectious Diseases. Clin. Microbiol. Rev. 2010, 23, 550–576. [Google Scholar] [CrossRef] [PubMed]
- Anders, C.K.; Johnson, R.; Litton, J.; Phillips, M.; Bleyer, A. Breast Cancer Before Age 40 Years. Semin. Oncol. 2009, 36, 237–249. [Google Scholar] [CrossRef]
- Cardoso, F.; Kyriakides, S.; Ohno, S.; Penault-Llorca, F.; Poortmans, P.; Rubio, I.T.; Zackrisson, S.; Senkus, E.; ESMO Guidelines Committee. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-updagger. Ann. Oncol. 2019, 30, 1194–1220. [Google Scholar] [CrossRef] [PubMed]
- Dogra, A.; Mehta, A.; Doval, D. Are Basal-Like and Non-Basal-Like Triple-Negative Breast Cancers Really Different? J. Oncol. 2020, 2020, 4061063. [Google Scholar] [CrossRef] [PubMed]
- Ginsburg, O.; Yip, C.; Brooks, A.; Cabanes, A.; Caleffi, M.; Yataco, J.A.D.; Gyawali, B.; McCormack, V.; de Anderson, M.M.; Mehrotra, R.; et al. Breast cancer early detection: A phased approach to implementation. Cancer 2020, 126 (Suppl. S10), 2379–2393. [Google Scholar] [CrossRef]
- Ingenito, F.; Roscigno, G.; Affinito, A.; Nuzzo, S.; Scognamiglio, I.; Quintavalle, C.; Condorelli, G. The Role of Exo-miRNAs in Cancer: A Focus on Therapeutic and Diagnostic Applications. Int. J. Mol. Sci. 2019, 20, 4687. [Google Scholar] [CrossRef]
Exosomal Markers | Body Fluid | Isolation/Detection Method | Ref. |
---|---|---|---|
CD24 | Serum | Ultracentrifugation/Western blot | [52] |
Survivin, Survivin-ΔEx3, Survivin-2B | Serum | ExoQuick/Western blot | [53] |
HER2 | Serum | Ultracentrifugation/Western blot | [54] |
EpCAM, HER2 | Plasma | Immunocapture on microfluidic chip/Immunofluorescence | [55] |
Glypican-1 | Serum | Ultracentrifugation/FACS | [56] |
Periostin | Plasma | Ultracentrifugation/Western blot | [57] |
CD47 | Serum | Total Exosome Isolation kit/Flow cytometry and ELISA | [58] |
Exo-miRNA Detection Kit | Pre-Amplification Step | Expertise in Bioinformatics Required | Normalization | Internal Controls | Cost- Effective |
---|---|---|---|---|---|
TaqMan MicroRNA array Human Pool A Card | Yes | No | The most suitable reference miR is selected from the experimental data. | One | Yes |
nCounter® miRNA Expression Panels | No | No | miRs for normalization are included by the manufacturer. | Included in the assay by the manufacturer | Yes |
Next generation sequencing | Yes. Library construction | Yes | Reads are mapped with a genome reference sequence (miRBase). | Included in the assay | No |
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. |
© 2023 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
Encarnación-Medina, J.; Godoy, L.; Matta, J.; Ortiz-Sánchez, C. Identification of Exo-miRNAs: A Summary of the Efforts in Translational Studies Involving Triple-Negative Breast Cancer. Cells 2023, 12, 1339. https://doi.org/10.3390/cells12091339
Encarnación-Medina J, Godoy L, Matta J, Ortiz-Sánchez C. Identification of Exo-miRNAs: A Summary of the Efforts in Translational Studies Involving Triple-Negative Breast Cancer. Cells. 2023; 12(9):1339. https://doi.org/10.3390/cells12091339
Chicago/Turabian StyleEncarnación-Medina, Jarline, Lenin Godoy, Jaime Matta, and Carmen Ortiz-Sánchez. 2023. "Identification of Exo-miRNAs: A Summary of the Efforts in Translational Studies Involving Triple-Negative Breast Cancer" Cells 12, no. 9: 1339. https://doi.org/10.3390/cells12091339
APA StyleEncarnación-Medina, J., Godoy, L., Matta, J., & Ortiz-Sánchez, C. (2023). Identification of Exo-miRNAs: A Summary of the Efforts in Translational Studies Involving Triple-Negative Breast Cancer. Cells, 12(9), 1339. https://doi.org/10.3390/cells12091339