miRNA in Molecular Diagnostics: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

miRNAs influence the expression of numerous proteins, including the expression of tumor suppressors and protooncogenes, thus becoming oncogenes and tumor suppressors themselves. As the same miRNA can have different targets in different tissues, its function will also be different in different types of tumors, depending on the intracellular milieu and the set of proteins for which its translation is modulated. Therefore, the same miRNA can act as a tumor suppressor and an oncogene in different tumors. Even in the same tumor, the same miRNA can be involved in regulation circles with feedback loops and potentially affect both tumor suppressors and oncogenes. As 50% of miRNA genes are located in regions associated with cancers, their expression is found to be deregulated in tumors. miRNAs were found to be members of signaling circuits, often involving also long non-coding RNAs (lnc RNAs) and circular RNAs (cRNAs).

  • miRNA
  • malignant tumors
  • viral infections

1. Introduction

As oncomirs and tumor suppressors, miRNAs mostly influence processes of cell proliferation and apoptosis. Examples of most investigated miRNAs in various malignant tumors and their targets are listed. [1][2][3][4][5][6][7][8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][73][74][75][76][77][78][79][80][81][82][83][84][85][86][87][88][89][90][91][92][93][94][95][96][97][98][99][100][101][102][103][104][105][106][107][108][109][110][111][112][113][114][115][116][117][118][119][120][121][122][123][124][125][126][127][128][129][130][131][132][133][134][135][136][137][138][139][140][141][142][143][144][145][146][147][148][149][150][151][152][153][154][155][156][157][158][159][160][161][162][163][164][165][166][167][168].
As illustrated on the example of osteosarcoma [169], miRNA can influence all hallmarks of cancer: cell cycle, proliferation control mechanisms, cell migration, metastasis and invasion, autophagy, apoptosis, senescence, and differentiation. They also affect resistance to chemotherapeutics, metabolism, and immune surveillance [47][111][133][141].
Specific changes in miRNA regulation can be found in all types of tumors, but detecting those that could be used as diagnostics markers is challenging, as they are cell- or tumor-specific. Currently, besides detecting one specific miRNA, often, a set of deregulated miRNAs is correlated with a certain type of tumor. For example, Sharma and Gupta analyzed miRNAs as potential biomarkers for the diagnosis and prognosis of different types of cancer and detected 723 dysregulated miRNAs in 16 types of tumors. Forty-three miRNAs were differentially expressed in six or more types of tumors [170].

2. miRNAs in Leukemia and Lymphoma

First miRNAs found to be directly involved in tumor development were detected in leukemia patients. The involvement in proliferation and apoptosis and their roles in the signaling loops in B cell differentiation are probably the best known and understood in hematopoietic cells and their malignancies [171]. Calin discovered that the deletion of 13q14 in chronic lymphocytic leukemia (CLL) coincided with a loss of miRNA15a and miRNA16-1 (miR15/16), and in nearly 70% of CLL patients, these miRNAs were found to be either absent or epigenetically downregulated [1]. Their deletion influenced the expression of antiapoptotic proteins BCL2 and MCL1, leading to survival and resistance to chemotherapeutics, as well as cyclin D1 expression and cell proliferation [1][2][3]. As their target is also p53, their deletion leads to the development of a specific CLL subtype. miR15/miR16 cluster downregulation was also found to be involved in other types of leukemia [4].
Another miRNA gene cluster for which its expression correlated with development of hematopoietic disorders was miR17-miR92 cluster, containing six miRNAs, produced from a polycistronic transcript [5]. Its amplification was found in B cell lymphoma, T cell leukemia, and some solid tumors [5][172]. Its locus is regulated by c-Myc, and its main targets are BIM, PTEN, p21, and p57 [173]. Its deregulation compromises apoptosis and increases proliferation, cell survival, resistance to chemotherapy, and BCR signaling [7][8][9].
Numerous miRNAs were further found to be involved in normal development and leukemogenesis in both B and T lymphocytes. One of them is miR34a, which acts as a tumor suppressor and is regulated by p53. Its main targets are FOXP1, ZAP-70, genes involved in apoptosis, such as BCL2, and genes regulating cell cycle progression and proliferation, such as BCL6, B MYB, CDK6, and AXL [10].
While FOXP1 is regulated by miR34a in pro/pre-B cells, miR150 regulates this gene in mature B cells. In pro-B cells, miR150 regulates Myb [11][171]. Its deletion increases BCR signaling and survival pathways involving PIK3AP1 and AKT2 and influences telomerase expression. By targeting CXCR4 it regulates mobilization and migration of mononuclear cells [12]. Its expression is downregulated in one CLL subtype and in different types of lymphomas.
miR155 can act as both an oncogene and a tumor suppressor in different steps of B cell development, and it is another example of different types of negative feedback loops present in the signaling regulations of miRNAs [171]. It is regulated by BCR activation, its downstream targets are transcription factor Pu.1 and AID, which are involved in immunoglobulin somatic hypermutation [13], and it regulates Akt signaling, proliferation, motility, and the modulation of TGFβ pathways [14]. miR181b also targets AID, in addition to Bcl2, MCL1 and TCL1, and Akt kinases coactivator, thus influencing apoptosis, cell survival, and differentiation. This miRNA is downregulated in CLL, and its levels can correlate with disease progression in the samples of the same patient [15][16].

3. miRNA in Brain Tumors

miRNA in glioblastoma and other brain tumors were analyzed by several research groups, detecting characteristic sets of 5–10 up- and downregulated miRNAs as specific signatures that correlated with patients’ survival. These miRNAs influence MAPK, PI3K/Akt, mTOR, and Wnt signaling pathways and deregulate apoptosis and the control of proliferation [20][29]. Among miRNAs, the most analyzed miRNA in these tumor types is miR7, and it is involved in neural cell differentiation and acts as a tumor suppressor. It mainly influences targets in Akt and MAP kinase pathways, and its downregulation increases proliferation, survival, and inhibits apoptosis [21][174]. Other miRNAs often involved in glioblastoma development are miR21, miR221, and miR181 [22][23][29]. miR21 overexpression leads to the inhibition of apoptosis and increases in cell proliferation [22][23][175]. Other mentioned miRNAs are involved in PI3K/Akt regulation, Notch and p53 signaling, and DNA repair [27][29][35].

4. miRNA in Lung Cancer

miR21 [37], miR148 [50], and miR205 [38] are among the most investigated miRNAs in lung cancer, but numerous other miRNAs were also found to be deregulated in this type of tumor. Several sets of miRNAs with altered expression were also identified in lung adenocarcinoma, in addition to observing differentially expressed miRNAs in different types of lung cancer [176][177][178][179]. It was found that EGFR mutation in lung cancer cells leads to changes in the expression of 17 miRNAs, including the miR17-92 cluster [52]. These miRNAs influence proliferation, survival, resistance to chemotherapy and apoptosis, and migration and cancer cell stemness [46][180]. The main targets of deregulated miRNAs in lung tumors are Ras and Myc pathways, PTEN and PI3K/Akt signaling leading to cell proliferation, the p53 pathway, and others influencing resistance to chemotherapeutics and apoptosis [38][42][43][44]. Cell migration, proliferation, and resistance to chemotherapeutics are influenced by the interaction of miRNA with HIF and TGFβ pathway elements [36][49][131]. Other processes regulated by miRNAs in lungs are metabolism and glycolysis, as well as epithelial–mesenchymal transition (EMT) [44][46][49][181][182].

5. miRNA in Breast Carcinoma

Numerous miRNAs and signatures of deregulated miRNA were detected in breast cancer [183][184]. Among those, the most investigated are miR125b, miR145, miR21, miR155, and miR205 [54][55][185]. The main targets of deregulated miRNAs are molecules involved in MAP/AKT/STAT3 signaling pathways and those regulating cell proliferation, epithelial–mesenchymal transition, angiogenesis by targeting VEGFA, cell stemness, and resistance to chemotherapy [34][53][56][57][58][59][186].

6. miRNA in Bladder and Renal Carcinoma

In bladder cancer, deregulated miRNAs often include miR34a, miR21, and miR222, and several miRNAs are linked to migration and invasion [69][70][71]. Their targets are β catenin, CDK2, E cadherin, as well as integrin α5, influencing resistance to chemotherapy [67][73][74].
In renal cancer tissues, the main targets of deregulated miRNAs include proteins participating in proliferation, such as those in Akt and Wnt signaling, migration, invasion, and EMT [72][77][78][80][187].

7. miRNA in Colon, Hepatocellular and Gastric Carcinoma

In colon cancer, deregulated miRNAs, including miR200c, miR145, miR181, miR101, and miR21, mainly interfere with cell proliferation and migration, apoptosis, Wnt/β-catenin, and MAPK pathways [81][83][84][87][88]. In this entity, specific sets of miRNAs with prognostic and diagnostic potential were detected [188].
The most significant miRNAs involved in signaling circuits in hepatocellular carcinoma are those regulating the PI3K/Akt pathway, cell proliferation, apoptosis, invasion, EMT, and glucose metabolism [90][92][93][94][95]. They mainly act as tumor suppressors [99][189][190].
In gastric carcinoma, miRNAs also regulate cell proliferation and migration by targeting PTEN and EGFR, as well as MAPK pathways, and EZH2, which participates in chromatin remodeling [99][100][102]. Numerous miRNAs in this disease are related to resistance to apoptosis through the regulation of Bcl2 or other members of its family, angiogenesis, and resistance to chemotherapy [105][106][107][191].
In pancreatic cancer, miRNAs regulate EMT through TGFβ signaling, as well as processes of invasion and the inhibition of apoptosis [108][109].

8. miRNA in Cervical Carcinoma, Testicular Tumors, and Prostate Cancer

In cervical carcinoma, miRNAs promote tumor proliferation, migration, invasion, and influence apoptosis and chemoresistance. Examples are miR21, which influences Akt/mTOR pathway, proliferation, growth, and EMT [113]; miR375, which targets E-cadherin; and miR138, which targets EZH2, influencing chromatin remodeling [114][192]. It was observed that viral proteins E6 and E7 increase the expression of miR18a [110], influencing Hippo signaling, in human papilloma virus (HPV)-associated cervical carcinoma.
In prostate cancer, miRNAs influence proliferation, apoptosis, migration and invasion. The main targets are Akt and MAPK pathways and HIF and VEGF pathways [114][119][123][124].
In different types of testicular germ cell tumors, several miRNAs are differently expressed and vary from low expression in teratoma, medium expression in seminoma, and high expression in embryonal carcinoma. The main deregulated miRNAs are miR199-214, influencing tumor metabolism through epigenetic regulators, miR371-373 influencing p53 pathway, cell cycle regulation, Wnt/β-catenin signaling, and senescence; and miR223, influencing apoptosis and cell growth through FBXW7 [125][127][128][132][133]. Other miRNA targets are cell cycle regulators, members of the p53 pathway involved in apoptosis regulation, DNA damage sensitivity, cell differentiation, and lactate metabolism [127][131][193][194].

9. miRNA in Skin Tumors

An analysis of metastatic melanoma revealed 44 miRNAs acting as tumor suppressors and 23 as oncomirs [195]. Some of those miRNAs control the expression of MITF, transcription factor involved in differentiation, proliferation, and the survival of melanocytes, cell motility, and invasiveness [196]. Numerous miRNAs control MITF directly, and others control MITF by targeting signaling pathways regulating its expression, such as Wnt and MAP signaling. Furthermore, some miRNAs regulate cell survival and take part in chromatin remodulation [135]. Developments in invasive melanoma are linked to melanoma phenotype switching when a highly proliferative state is exchanged for invasive states characterized by its migration ability. In this state, MITF levels decrease. Phenotypic changes have similarities to EMT, and numerous miRNAs involved in the regulation of migration are deregulated. High MITF expressions also correlate with resistance to chemotherapy, and nearly 20 tumor suppressors and oncomirs are linked to this process, with most of them targeting MAP kinase and PI3K and EMT pathways [138][139]. miRNAs in melanoma also regulate escape from immune surveillance [141].
In cutaneous squamous cell carcinoma, miRNAs influence cell proliferation, invasion, and migration; and inhibit apoptosis and differentiation by targeting PTEN, members of MAP kinase, and cMyc pathways [143][144][146].

10. miRNA in Other Tumors

Aplastic thyroid cancer is a highly invasive thyroid tumor that is fast growing and resistant to chemotherapy. miRNAs influence cell proliferation, invasion and EMT, cell adhesion, differentiation, and cell stemness by targeting PTEN, CDKI, NFκB, TGFβ, Wnt pathway, and ZEB, which are proteins involved in autophagy, apoptosis, and chromatin modulation [152][154][155][158][160][197]. In a rare medullary thyroid carcinoma, specific sets of deregulated miRNAs were detected [155][162].
In osteosarcoma, most deregulated miRNAs are linked to cell proliferation and migration: targeting β-catenin and MAP kinases pathways [164][165][166]. There are miRNAs that act as oncomirs and tumor suppressors depending on the intracellular milieu of different osteosarcomas [167][168][169].

This entry is adapted from the peer-reviewed paper 10.3390/bioengineering9090459

References

  1. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529.
  2. Calin, G.A.; Cimmino, A.; Fabbri, M.; Ferracin, M.; Wojcik, S.E.; Shimizu, M.; Taccioli, C.; Zanesi, N.; Garzon, R.; Aqeilan, R.I.; et al. MiR-15a and miR-16-1 cluster functions in human leukemia. Proc. Natl. Acad. Sci. USA 2008, 105, 5166–5171.
  3. Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949.
  4. Lovat, F.; Fassan, M.; Sacchi, D.; Ranganathan, P.; Palamarchuk, A.; Bill, M.; Karunasiri, M.; Gasparini, P.; Nigita, G.; Distefano, R.; et al. Knockout of both miR-15/16 loci induces acute myeloid leukemia. Proc. Natl. Acad. Sci. USA 2018, 115, 13069–13074.
  5. He, L.; Thomson, J.M.; Hemann, M.T.; Hernando-Monge, E.; Mu, D.; Goodson, S.; Powers, S.; Cordon-Cardo, C.; Lowe, S.W.; Hannon, G.J.; et al. A microRNA polycistron as a potential human oncogene. Nature 2005, 435, 828–833.
  6. Xiao, C.; Srinivasan, L.; Calado, D.P.; Patterson, H.C.; Zhang, B.; Wang, J.; Henderson, J.M.; Kutok, J.L.; Rajewsky, K. Lymphoproliferative disease and autoimmunity in mice with increased miR-17-92 expression in lymphocytes. Nat. Immunol. 2008, 9, 405–414.
  7. Psathas, J.N.; Doonan, P.J.; Raman, P.; Freedman, B.D.; Minn, A.J.; Thomas-Tikhonenko, A. The Myc-miR-17-92 axis amplifies B-cell receptor signaling via inhibition of ITIM proteins: A novel lymphomagenic feed-forward loop. Blood 2013, 122, 4220–4229.
  8. Scherr, M.; Elder, A.; Battmer, K.; Barzan, D.; Bomken, S.; Ricke-Hoch, M.; Schröder, A.; Venturini, L.; Blair, H.J.; Vormoor, J.; et al. Differential expression of miR-17~92 identifies BCL2 as a therapeutic target in BCR-ABL-positive B-lineage acute lymphoblastic leukemia. Leukemia 2014, 28, 554–565.
  9. Jin, H.Y.; Oda, H.; Lai, M.; Skalsky, R.L.; Bethel, K.; Shepherd, J.; Kang, S.G.; Liu, W.H.; Sabouri-Ghomi, M.; Cullen, B.R.; et al. MicroRNA-17~92 plays a causative role in lymphomagenesis by coordinating multiple oncogenic pathways. EMBO J. 2013, 32, 2377–2391.
  10. Cerna, K.; Oppelt, J.; Chochola, V.; Musilova, K.; Seda, V.; Pavlasova, G.; Radova, L.; Arigoni, M.; Calogero, R.A.; Benes, V.; et al. MicroRNA miR-34a downregulates FOXP1 during DNA damage response to limit BCR signalling in chronic lymphocytic leukaemia B cells. Leukemia 2019, 33, 403–414.
  11. Mraz, M.; Chen, L.; Rassenti, L.Z.; Ghia, E.M.; Li, H.; Jepsen, K.; Smith, E.N.; Messer, K.; Frazer, K.A.; Kipps, T.J. miR-150 influences B-cell receptor signaling in chronic lymphocytic leukemia by regulating expression of GAB1 and FOXP1. Blood 2014, 124, 84–95.
  12. Tano, N.; Kim, H.W.; Ashraf, M. microRNA-150 regulates mobilization and migration of bone marrow-derived mononuclear cells by targeting Cxcr4. PLoS ONE 2011, 6, e23114.
  13. Vigorito, E.; Perks, K.L.; Abreu-Goodger, C.; Bunting, S.; Xiang, Z.; Kohlhaas, S.; Das, P.P.; Miska, E.A.; Rodriguez, A.; Bradley, A.; et al. microRNA-155 regulates the generation of immunoglobulin class-switched plasma cells. Immunity 2007, 27, 847–859.
  14. Eis, P.S.; Tam, W.; Sun, L.; Chadburn, A.; Li, Z.; Gomez, M.F.; Lund, E.; Dahlberg, J.E. Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proc. Natl. Acad. Sci. USA 2005, 102, 3627–3632.
  15. Calin, G.A.; Ferracin, M.; Cimmino, A.; Di Leva, G.; Shimizu, M.; Wojcik, S.E.; Iorio, M.V.; Visone, R.; Sever, N.I.; Fabbri, M.; et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N. Engl. J. Med. 2005, 353, 1793–1801.
  16. Pekarsky, Y.; Santanam, U.; Cimmino, A.; Palamarchuk, A.; Efanov, A.; Maximov, V.; Volinia, S.; Alder, H.; Liu, C.G.; Rassenti, L.; et al. Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res. 2006, 66, 11590–11593.
  17. Medina, P.P.; Nolde, M.; Slack, F.J. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature 2010, 467, 86–90.
  18. Valeri, N.; Gasparini, P.; Braconi, C.; Paone, A.; Lovat, F.; Fabbri, M.; Sumani, K.M.; Alder, H.; Amadori, D.; Patel, T.; et al. MicroRNA-21 induces resistance to 5-fluorouracil by down-regulating human DNA MutS homolog 2 (hMSH2). Proc. Natl. Acad. Sci. USA 2010, 107, 21098–21103.
  19. Zhao, J.J.; Lin, J.; Lwin, T.; Yang, H.; Guo, J.; Kong, W.; Dessureault, S.; Moscinski, L.C.; Rezania, D.; Dalton, W.S.; et al. microRNA expression profile and identification of miR-29 as a prognostic marker and pathogenetic factor by targeting CDK6 in mantle cell lymphoma. Blood 2010, 115, 2630–2639.
  20. Basso, J.; Paggi, M.G.; Fortuna, A.; Vitorino, C.; Vitorino, R. Deciphering specific miRNAs in brain tumors: A 5-miRNA signature in glioblastoma. Mol. Genet. Genomics 2022, 297, 507–521.
  21. Kefas, B.; Godlewski, J.; Comeau, L.; Li, Y.; Abounader, R.; Hawkinson, M.; Lee, J.; Fine, H.; Chiocca, E.A.; Lawler, S.; et al. microRNA-7 inhibits the epidermal growth factor receptor and the Akt pathway and is down-regulated in glioblastoma. Cancer Res. 2008, 68, 3566–3572.
  22. Papagiannakopoulos, T.; Shapiro, A.; Kosik, K.S. MicroRNA-21 targets a network of key tumor-suppressive pathways in glioblastoma cells. Cancer Res. 2008, 68, 8164–8172.
  23. Chan, J.A.; Krichevsky, A.M.; Kosik, K.S. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005, 65, 6029–6033.
  24. Costa, P.M.; Cardoso, A.L.; Pereira de Almeida, L.F.; Bruce, J.N.; Canoll, P.; Pedroso de Lima, M.C. PDGF-B-mediated downregulation of miR-21: New insights into PDGF signaling in glioblastoma. Hum. Mol. Genet. 2012, 21, 5118–5130.
  25. Godlewski, J.; Nowicki, M.O.; Bronisz, A.; Nuovo, G.; Palatini, J.; De Lay, M.; Van Brocklyn, J.; Ostrowski, M.C.; Chiocca, E.A.; Lawler, S.E. MicroRNA-451 regulates LKB1/AMPK signaling and allows adaptation to metabolic stress in glioma cells. Mol. Cell 2010, 37, 620–632.
  26. Ogawa, D.; Ansari, K.; Nowicki, M.O.; Salińska, E.; Bronisz, A.; Godlewski, J. MicroRNA-451 Inhibits Migration of Glioblastoma while Making It More Susceptible to Conventional Therapy. Noncoding RNA 2019, 5, 25.
  27. Kefas, B.; Comeau, L.; Erdle, N.; Montgomery, E.; Amos, S.; Purow, B. Pyruvate kinase M2 is a target of the tumor-suppressive microRNA-326 and regulates the survival of glioma cells. Neuro Oncol. 2010, 12, 1102–1112.
  28. Li, Y.; Guessous, F.; Zhang, Y.; Dipierro, C.; Kefas, B.; Johnson, E.; Marcinkiewicz, L.; Jiang, J.; Yang, Y.; Schmittgen, T.D.; et al. MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes. Cancer Res. 2009, 69, 7569–7576.
  29. Ciafrè, S.A.; Galardi, S.; Mangiola, A.; Ferracin, M.; Liu, C.G.; Sabatino, G.; Negrini, M.; Maira, G.; Croce, C.M.; Farace, M.G. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem. Biophys. Res. Commun. 2005, 334, 1351–1358.
  30. Godlewski, J.; Nowicki, M.O.; Bronisz, A.; Williams, S.; Otsuki, A.; Nuovo, G.; Raychaudhury, A.; Newton, H.B.; Chiocca, E.A.; Lawler, S. Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal. Cancer Res. 2008, 68, 9125–9130.
  31. Peruzzi, P.; Bronisz, A.; Nowicki, M.O.; Wang, Y.; Ogawa, D.; Price, R.; Nakano, I.; Kwon, C.H.; Hayes, J.; Lawler, S.E.; et al. MicroRNA-128 coordinately targets Polycomb Repressor Complexes in glioma stem cells. Neuro Oncol. 2013, 15, 1212–1224.
  32. Conti, A.; Aguennouz, M.; La Torre, D.; Tomasello, C.; Cardali, S.; Angileri, F.F.; Maio, F.; Cama, A.; Germanò, A.; Vita, G.; et al. miR-21 and 221 upregulation and miR-181b downregulation in human grade II-IV astrocytic tumors. J. Neurooncol. 2009, 93, 325–332.
  33. Chen, G.; Zhu, W.; Shi, D.; Lv, L.; Zhang, C.; Liu, P.; Hu, W. MicroRNA-181a sensitizes human malignant glioma U87MG cells to radiation by targeting Bcl-2. Oncol. Rep. 2010, 23, 997–1003.
  34. Wang, H.; Tao, T.; Yan, W.; Feng, Y.; Wang, Y.; Cai, J.; You, Y.; Jiang, T.; Jiang, C. Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma. Sci. Rep. 2015, 5, 13072.
  35. Medina, R.; Zaidi, S.K.; Liu, C.G.; Stein, J.L.; van Wijnen, A.J.; Croce, C.M.; Stein, G.S. MicroRNAs 221 and 222 bypass quiescence and compromise cell survival. Cancer Res. 2008, 68, 2773–2780.
  36. Lin, L.; Tu, H.B.; Wu, L.; Liu, M.; Jiang, G.N. MicroRNA-21 Regulates Non-Small Cell Lung Cancer Cell Invasion and Chemo-Sensitivity through SMAD7. Cell. Physiol. Biochem. 2016, 38, 2152–2162.
  37. Seike, M.; Goto, A.; Okano, T.; Bowman, E.D.; Schetter, A.J.; Horikawa, I.; Mathe, E.A.; Jen, J.; Yang, P.; Sugimura, H.; et al. MiR-21 is an EGFR-regulated anti-apoptotic factor in lung cancer in never-smokers. Proc. Natl. Acad. Sci. USA 2009, 106, 12085–12090.
  38. Markou, A.; Tsaroucha, E.G.; Kaklamanis, L.; Fotinou, M.; Georgoulias, V.; Lianidou, E.S. Prognostic value of mature microRNA-21 and microRNA-205 overexpression in non-small cell lung cancer by quantitative real-time RT-PCR. Clin. Chem. 2008, 54, 1696–1704.
  39. Cai, J.; Fang, L.; Huang, Y.; Li, R.; Yuan, J.; Yang, Y.; Zhu, X.; Chen, B.; Wu, J.; Li, M. miR-205 targets PTEN and PHLPP2 to augment AKT signaling and drive malignant phenotypes in non-small cell lung cancer. Cancer Res. 2013, 73, 5402–5415.
  40. Larzabal, L.; de Aberasturi, A.L.; Redrado, M.; Rueda, P.; Rodriguez, M.J.; Bodegas, M.E.; Montuenga, L.M.; Calvo, A. TMPRSS4 regulates levels of integrin α5 in NSCLC through miR-205 activity to promote metastasis. Br. J. Cancer 2014, 110, 764–774.
  41. Kumar, M.S.; Erkeland, S.J.; Pester, R.E.; Chen, C.Y.; Ebert, M.S.; Sharp, P.A.; Jacks, T. Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proc. Natl. Acad. Sci. USA 2008, 105, 3903–3908.
  42. Johnson, S.M.; Grosshans, H.; Shingara, J.; Byrom, M.; Jarvis, R.; Cheng, A.; Labourier, E.; Reinert, K.L.; Brown, D.; Slack, F.J. RAS is regulated by the let-7 microRNA family. Cell 2005, 120, 635–647.
  43. Kanthaje, S.; Baikunje, N.; Kandal, I.; Ratnacaram, C.K. Repertoires of MicroRNA-30 family as gate-keepers in lung cancer. Front. Biosci. 2021, 13, 141–156.
  44. Song, K.; Jiang, Y.; Zhao, Y.; Xie, Y.; Zhou, J.; Yu, W.; Wang, Q. Members of the miR-30 family inhibit the epithelial-to-mesenchymal transition of non-small-cell lung cancer cells by suppressing XB130 expression levels. Oncol. Lett. 2020, 20, 68.
  45. He, Y.; Liu, H.; Jiang, L.; Rui, B.; Mei, J.; Xiao, H. miR-26 Induces Apoptosis and Inhibits Autophagy in Non-small Cell Lung Cancer Cells by Suppressing TGF-β1-JNK Signaling Pathway. Front. Pharmacol. 2019, 9, 1509.
  46. Hu, J.; Qiu, M.; Jiang, F.; Zhang, S.; Yang, X.; Wang, J.; Xu, L.; Yin, R. MiR-145 regulates cancer stem-like properties and epithelial-to-mesenchymal transition in lung adenocarcinoma-initiating cells. Tumour Biol. 2014, 35, 8953–8961.
  47. Xu, Y.J.; Wei, R.S.; Li, X.H.; Li, Q.; Yu, J.R.; Zhuang, X.F. MiR-421 promotes lipid metabolism by targeting PTEN via activating PI3K/AKT/mTOR pathway in non-small cell lung cancer. Epigenomics, 2022; Online ahead of print.
  48. Li, Y.; Zhao, L.; Qi, Y.; Yang, X. MicroRNA-214 upregulates HIF-1α and VEGF by targeting ING4 in lung cancer cells. Mol. Med. Rep. 2019, 19, 4935–4945.
  49. Liu, C.; Luo, J.; Zhao, Y.T.; Wang, Z.Y.; Zhou, J.; Huang, S.; Huang, J.N.; Long, H.X.; Zhu, B. TWIST1 upregulates miR-214 to promote epithelial-to-mesenchymal transition and metastasis in lung adenocarcinoma. Int. J. Mol. Med. 2018, 42, 461–470.
  50. Joshi, P.; Jeon, Y.J.; Laganà, A.; Middleton, J.; Secchiero, P.; Garofalo, M.; Croce, C.M. MicroRNA-148a reduces tumorigenesis and increases TRAIL-induced apoptosis in NSCLC. Proc. Natl. Acad. Sci. USA 2015, 112, 8650–8655.
  51. Lin, H.Y.; Chiang, C.H.; Hung, W.C. STAT3 upregulates miR-92a to inhibit RECK expression and to promote invasiveness of lung cancer cells. Br. J. Cancer 2013, 109, 731–738.
  52. Zhang, X.; Li, Y.; Qi, P.; Ma, Z. Biology of MiR-17-92 Cluster and Its Progress in Lung Cancer. Int. J. Med. Sci. 2018, 15, 1443–1448.
  53. Ninio-Many, L.; Hikri, E.; Burg-Golani, T.; Stemmer, S.M.; Shalgi, R.; Ben-Aharon, I. miR-125a Induces HER2 Expression and Sensitivity to Trastuzumab in Triple-Negative Breast Cancer Lines. Front. Oncol. 2020, 10, 191.
  54. Nandy, S.B.; Arumugam, A.; Subramani, R.; Pedroza, D.; Hernandez, K.; Saltzstein, E.; Lakshmanaswamy, R. MicroRNA-125a influences breast cancer stem cells by targeting leukemia inhibitory factor receptor which regulates the Hippo signaling pathway. Oncotarget 2015, 6, 17366–17378.
  55. Iorio, M.V.; Ferracin, M.; Liu, C.G.; Veronese, A.; Spizzo, R.; Sabbioni, S.; Magri, E.; Pedriali, M.; Fabbri, M.; Campiglio, M.; et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005, 65, 7065–7070.
  56. Adachi, R.; Horiuchi, S.; Sakurazawa, Y.; Hasegawa, T.; Sato, K.; Sakamaki, T. ErbB2 down-regulates microRNA-205 in breast cancer. Biochem. Biophys. Res. Commun. 2011, 411, 804–808.
  57. Elgamal, O.A.; Park, J.K.; Gusev, Y.; Azevedo-Pouly, A.C.; Jiang, J.; Roopra, A.; Schmittgen, T.D. Tumor suppressive function of mir-205 in breast cancer is linked to HMGB3 regulation. PLoS ONE 2013, 8, e76402.
  58. Liang, H.; Xiao, J.; Zhou, Z.; Wu, J.; Ge, F.; Li, Z.; Zhang, H.; Sun, J.; Li, F.; Liu, R.; et al. Hypoxia induces miR-153 through the IRE1α-XBP1 pathway to fine tune the HIF1α/VEGFA axis in breast cancer angiogenesis. Oncogene 2018, 37, 1961–1975.
  59. Yu, X.; Luo, A.; Liu, Y.; Wang, S.; Li, Y.; Shi, W.; Liu, Z.; Qu, X. MiR-214 increases the sensitivity of breast cancer cells to tamoxifen and fulvestrant through inhibition of autophagy. Mol. Cancer 2015, 14, 208.
  60. Wang, F.; Lv, P.; Liu, X.; Zhu, M.; Qiu, X. microRNA-214 enhances the invasion ability of breast cancer cells by targeting p53. Int. J. Mol. Med. 2015, 35, 1395–1402.
  61. Frankel, L.B.; Christoffersen, N.R.; Jacobsen, A.; Lindow, M.; Krogh, A.; Lund, A.H. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J. Biol. Chem. 2008, 283, 1026–1033.
  62. Yu, F.; Yao, H.; Zhu, P.; Zhang, X.; Pan, Q.; Gong, C.; Huang, Y.; Hu, X.; Su, F.; Lieberman, J.; et al. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell 2007, 131, 1109–1123.
  63. Ma, L.; Teruya-Feldstein, J.; Weinberg, R.A. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 2007, 449, 682–688.
  64. Camps, C.; Buffa, F.M.; Colella, S.; Moore, J.; Sotiriou, C.; Sheldon, H.; Harris, A.L.; Gleadle, J.M.; Ragoussis, J. hsa-miR-210 Is induced by hypoxia and is an independent prognostic factor in breast cancer. Clin. Cancer Res. 2008, 14, 1340–1348.
  65. Hu, Y.; Qiu, Y.; Yagüe, E.; Ji, W.; Liu, J.; Zhang, J. miRNA-205 targets VEGFA and FGF2 and regulates resistance to chemotherapeutics in breast cancer. Cell Death Dis. 2016, 7, e2291.
  66. Reddy, S.D.; Ohshiro, K.; Rayala, S.K.; Kumar, R. MicroRNA-7, a homeobox D10 target, inhibits p21-activated kinase 1 and regulates its functions. Cancer Res. 2008, 68, 8195–8200.
  67. Zhang, Q.; Wang, C.; Miao, S.; Li, C.; Chen, Z.; Li, F. Enhancing E-cadherin expression via promoter-targeted miR-373 suppresses bladder cancer cells growth and metastasis. Oncotarget 2017, 8, 93969–93983.
  68. Wang, Y.; Xu, Z.; Wang, X. miRNA-373 promotes urinary bladder cancer cell proliferation, migration and invasion through upregulating epidermal growth factor receptor. Exp. Ther. Med. 2019, 17, 1190–1195.
  69. Ohno, R.; Uozaki, H.; Kikuchi, Y.; Kumagai, A.; Aso, T.; Watanabe, M.; Watabe, S.; Muto, S.; Yamaguchi, R. Both cancerous miR-21 and stromal miR-21 in urothelial carcinoma are related to tumour progression. Histopathology 2016, 69, 993–999.
  70. Andrew, A.S.; Marsit, C.J.; Schned, A.R.; Seigne, J.D.; Kelsey, K.T.; Moore, J.H.; Perreard, L.; Karagas, M.R.; Sempere, L.F. Expression of tumor suppressive microRNA-34a is associated with a reduced risk of bladder cancer recurrence. Int. J. Cancer 2015, 137, 1158–1166.
  71. Wszolek, M.F.; Rieger-Christ, K.M.; Kenney, P.A.; Gould, J.J.; Silva Neto, B.; Lavoie, A.K.; Logvinenko, T.; Libertino, J.A.; Summerhayes, I.C. A MicroRNA expression profile defining the invasive bladder tumor phenotype. Urol. Oncol. 2011, 29, 794–801.
  72. Gottardo, F.; Liu, C.G.; Ferracin, M.; Calin, G.A.; Fassan, M.; Bassi, P.; Sevignani, C.; Byrne, D.; Negrini, M.; Pagano, F.; et al. Micro-RNA profiling in kidney and bladder cancers. Urol. Oncol. 2007, 25, 387–392.
  73. Mao, X.W.; Xiao, J.Q.; Li, Z.Y.; Zheng, Y.C.; Zhang, N. Effects of microRNA-135a on the epithelial-mesenchymal transition, migration and invasion of bladder cancer cells by targeting GSK3β through the Wnt/β-catenin signaling pathway. Exp. Mol. Med. 2018, 50, e429.
  74. Xu, T.; Qin, L.; Zhu, Z.; Wang, X.; Liu, Y.; Fan, Y.; Zhong, S.; Wang, X.; Zhang, X.; Xia, L.; et al. MicroRNA-31 functions as a tumor suppressor and increases sensitivity to mitomycin-C in urothelial bladder cancer by targeting integrin α5. Oncotarget 2016, 7, 27445–27457.
  75. Dey, N.; Das, F.; Ghosh-Choudhury, N.; Mandal, C.C.; Parekh, D.J.; Block, K.; Kasinath, B.S.; Abboud, H.E.; Choudhury, G.G. microRNA-21 governs TORC1 activation in renal cancer cell proliferation and invasion. PLoS ONE 2012, 7, e37366.
  76. Xu, X.; Wu, J.; Li, S.; Hu, Z.; Xu, X.; Zhu, Y.; Liang, Z.; Wang, X.; Lin, Y.; Mao, Y.; et al. Downregulation of microRNA-182-5p contributes to renal cell carcinoma proliferation via activating the AKT/FOXO3a signaling pathway. Mol. Cancer 2014, 13, 109.
  77. Sun, P.; Wang, L.; Lu, Y.; Liu, Y.; Li, L.; Yin, L.; Zhang, C.; Zhao, W.; Shen, B.; Xu, W. MicroRNA-195 targets VEGFR2 and has a tumor suppressive role in ACHN cells via PI3K/Akt and Raf/MEK/ERK signaling pathways. Int. J. Oncol. 2016, 49, 1155–1163.
  78. Liu, F.; Wu, L.; Wang, A.; Xu, Y.; Luo, X.; Liu, X.; Hua, Y.; Zhang, D.; Wu, S.; Lin, T.; et al. MicroRNA-138 attenuates epithelial-to-mesenchymal transition by targeting SOX4 in clear cell renal cell carcinoma. Am. J. Transl. Res. 2017, 9, 3611–3622.
  79. Lu, J.; Wei, J.H.; Feng, Z.H.; Chen, Z.H.; Wang, Y.Q.; Huang, Y.; Fang, Y.; Liang, Y.P.; Cen, J.J.; Pan, Y.H.; et al. miR-106b-5p promotes renal cell carcinoma aggressiveness and stem-cell-like phenotype by activating Wnt/β-catenin signalling. Oncotarget 2017, 8, 21461–21471.
  80. Hu, G.; Lai, P.; Liu, M.; Xu, L.; Guo, Z.; Liu, H.; Li, W.; Wang, G.; Yao, X.; Zheng, J.; et al. miR-203a regulates proliferation, migration, and apoptosis by targeting glycogen synthase kinase-3β in human renal cell carcinoma. Tumour Biol. 2014, 35, 11443–11453.
  81. Strillacci, A.; Griffoni, C.; Sansone, P.; Paterini, P.; Piazzi, G.; Lazzarini, G.; Spisni, E.; Pantaleo, M.A.; Biasco, G.; Tomasi, V. MiR-101 downregulation is involved in cyclooxygenase-2 overexpression in human colon cancer cells. Exp. Cell Res. 2009, 315, 1439–1447.
  82. Ding, L.; Yu, L.L.; Han, N.; Zhang, B.T. miR-141 promotes colon cancer cell proliferation by inhibiting MAP2K4. Oncol. Lett. 2017, 13, 1665–1671.
  83. Tian, Y.; Pan, Q.; Shang, Y.; Zhu, R.; Ye, J.; Liu, Y.; Zhong, X.; Li, S.; He, Y.; Chen, L.; et al. MicroRNA-200 (miR-200) cluster regulation by achaete scute-like 2 (Ascl2): Impact on the epithelial-mesenchymal transition in colon cancer cells. J. Biol. Chem. 2014, 289, 36101–36115.
  84. Wang, P.; Zou, F.; Zhang, X.; Li, H.; Dulak, A.; Tomko, R.J., Jr.; Lazo, J.S.; Wang, Z.; Zhang, L.; Yu, J. microRNA-21 negatively regulates Cdc25A and cell cycle progression in colon cancer cells. Cancer Res. 2009, 69, 8157–8165.
  85. Yu, Y.; Kanwar, S.S.; Patel, B.B.; Oh, P.S.; Nautiyal, J.; Sarkar, F.H.; Majumdar, A.P. MicroRNA-21 induces stemness by downregulating transforming growth factor beta receptor 2 (TGFβR2) in colon cancer cells. Carcinogenesis 2012, 33, 68–76.
  86. Christensen, L.L.; Tobiasen, H.; Holm, A.; Schepeler, T.; Ostenfeld, M.S.; Thorsen, K.; Rasmussen, M.H.; Birkenkamp-Demtroeder, K.; Sieber, O.M.; Gibbs, P.; et al. MiRNA-362-3p induces cell cycle arrest through targeting of E2F1, USF2 and PTPN1 and is associated with recurrence of colorectal cancer. Int. J. Cancer 2013, 133, 67–78.
  87. Wang, Z.; Zhang, X.; Yang, Z.; Du, H.; Wu, Z.; Gong, J.; Yan, J.; Zheng, Q. MiR-145 regulates PAK4 via the MAPK pathway and exhibits an antitumor effect in human colon cells. Biochem. Biophys. Res. Commun. 2012, 427, 444–449.
  88. Yang, X.; Sun, Y.; Zhang, Y.; Han, S. Downregulation of miR-181b inhibits human colon cancer cell proliferation by targeting CYLD and inhibiting the NF-κB signaling pathway. Int. J. Mol. Med. 2020, 46, 1755–1764.
  89. Chen, F.; Li, Z.; Zhou, H. Identification of prognostic miRNA biomarkers for predicting overall survival of colon adenocarcinoma and bioinformatics analysis: A study based on the Cancer Genome Atlas database. J. Cell. Biochem. 2019, 120, 9839–9849.
  90. Zhang, W.; Liu, Y.; Fu, Y.; Han, W.; Xu, H.; Wen, L.; Deng, Y.; Liu, K. Long non-coding RNA LINC00160 functions as a decoy of microRNA-132 to mediate autophagy and drug resistance in hepatocellular carcinoma via inhibition of PIK3R3. Cancer Lett 2020, 478, 22–33.
  91. Bao, C.; Li, Y.; Huan, L.; Zhang, Y.; Zhao, F.; Wang, Q.; Liang, L.; Ding, J.; Liu, L.; Chen, T.; et al. NF-κB signaling relieves negative regulation by miR-194 in hepatocellular carcinoma by suppressing the transcription factor HNF-1α. Sci. Signal. 2015, 8, ra75.
  92. Cui, G.; Wang, H.; Liu, W.; Xing, J.; Song, W.; Zeng, Z.; Liu, L.; Wang, H.; Wang, X.; Luo, H.; et al. Glycogen Phosphorylase B Is Regulated by miR101-3p and Promotes Hepatocellular Carcinoma Tumorigenesis. Front. Cell Dev. Biol. 2020, 8, 566494.
  93. Zhang, X.N.; Zhou, J.; Lu, X.J. The long noncoding RNA NEAT1 contributes to hepatocellular carcinoma development by sponging miR-485 and enhancing the expression of the STAT3. J. Cell. Physiol. 2018, 233, 6733–6741.
  94. Wang, Y.; Lee, A.T.; Ma, J.Z.; Wang, J.; Ren, J.; Yang, Y.; Tantoso, E.; Li, K.B.; Ooi, L.L.; Tan, P.; et al. Profiling microRNA expression in hepatocellular carcinoma reveals microRNA-224 up-regulation and apoptosis inhibitor-5 as a microRNA-224-specific target. J. Biol. Chem. 2008, 283, 13205–13215.
  95. Connolly, E.; Melegari, M.; Landgraf, P.; Tchaikovskaya, T.; Tennant, B.C.; Slagle, B.L.; Rogler, L.E.; Zavolan, M.; Tuschl, T.; Rogler, C.E. Elevated expression of the miR-17-92 polycistron and miR-21 in hepadnavirus-associated hepatocellular carcinoma contributes to the malignant phenotype. Am. J. Pathol. 2008, 173, 856–864.
  96. Kong, J.; Liu, X.; Li, X.; Wu, J.; Wu, N.; Chen, J.; Fang, F. miR-125/Pokemon auto-circuit contributes to the progression of hepatocellular carcinoma. Tumour Biol. 2016, 37, 511–519.
  97. Li, W.; Xie, L.; He, X.; Li, J.; Tu, K.; Wei, L.; Wu, J.; Guo, Y.; Ma, X.; Zhang, P.; et al. Diagnostic and prognostic implications of microRNAs in human hepatocellular carcinoma. Int. J. Cancer 2008, 123, 1616–1622.
  98. Murakami, Y.; Yasuda, T.; Saigo, K.; Urashima, T.; Toyoda, H.; Okanoue, T.; Shimotohno, K. Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene 2006, 25, 2537–2545.
  99. Yang, T.S.; Yang, X.H.; Wang, X.D.; Wang, Y.L.; Zhou, B.; Song, Z.S. MiR-214 regulate gastric cancer cell proliferation, migration and invasion by targeting PTEN. Cancer Cell Int. 2013, 13, 68.
  100. Carvalho, J.; van Grieken, N.C.; Pereira, P.M.; Sousa, S.; Tijssen, M.; Buffart, T.E.; Diosdado, B.; Grabsch, H.; Santos, M.A.; Meijer, G.; et al. Lack of microRNA-101 causes E-cadherin functional deregulation through EZH2 up-regulation in intestinal gastric cancer. J. Pathol. 2012, 228, 31–44.
  101. Ning, T.; Zhang, H.; Wang, X.; Li, S.; Zhang, L.; Deng, T.; Zhou, L.; Liu, R.; Wang, X.; Bai, M.; et al. miR-370 regulates cell proliferation and migration by targeting EGFR in gastric cancer. Oncol. Rep. 2017, 38, 384–392.
  102. Wang, Y.; Zhang, H.; Ge, S.; Fan, Q.; Zhou, L.; Li, H.; Bai, M.; Ning, T.; Liu, R.; Wang, X.; et al. Effects of miR-138-5p and miR-204-5p on the migration and proliferation of gastric cancer cells by targeting EGFR. Oncol. Rep. 2018, 39, 2624–2634.
  103. Kogo, R.; Mimori, K.; Tanaka, F.; Komune, S.; Mori, M. Clinical significance of miR-146a in gastric cancer cases. Clin. Cancer Res. 2011, 17, 4277–4284.
  104. Yao, Y.; Suo, A.L.; Li, Z.F.; Liu, L.Y.; Tian, T.; Ni, L.; Zhang, W.G.; Nan, K.J.; Song, T.S.; Huang, C. MicroRNA profiling of human gastric cancer. Mol. Med. Rep. 2009, 2, 963–970.
  105. Lu, Z.; Liu, M.; Stribinskis, V.; Klinge, C.M.; Ramos, K.S.; Colburn, N.H.; Li, Y. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene 2008, 27, 4373–4379.
  106. Tao, S.; Gu, J.; Wang, Q.; Zheng, L. Translational control of Bcl-2 promotes apoptosis of gastric carcinoma cells. BMC Cancer 2021, 21, 12.
  107. Zhou, J.; Chen, Q. Poor expression of microRNA-135b results in the inhibition of cisplatin resistance and proliferation and induces the apoptosis of gastric cancer cells through MST1-mediated MAPK signaling pathway. FASEB J. 2019, 33, 3420–3436.
  108. Mody, H.R.; Hung, S.W.; Pathak, R.K.; Griffin, J.; Cruz-Monserrate, Z.; Govindarajan, R. miR-202 Diminishes TGFβ Receptors and Attenuates TGFβ1-Induced EMT in Pancreatic Cancer. Mol. Cancer Res. 2017, 15, 1029–1039.
  109. Zhu, Z.; Xu, Y.; Zhao, J.; Liu, Q.; Feng, W.; Fan, J.; Wang, P. miR-367 promotes epithelial-to-mesenchymal transition and invasion of pancreatic ductal adenocarcinoma cells by targeting the Smad7-TGF-β signalling pathway. Br. J. Cancer 2015, 112, 1367–1375.
  110. Morgan, E.L.; Patterson, M.R.; Ryder, E.L.; Lee, S.Y.; Wasson, C.W.; Harper, K.L.; Li, Y.; Griffin, S.; Blair, G.E.; Whitehouse, A.; et al. MicroRNA-18a targeting of the STK4/MST1 tumour suppressor is necessary for transformation in HPV positive cervical cancer. PLoS Pathog. 2020, 16, e1008624.
  111. Shen, Y.; Zhou, J.; Li, Y.; Ye, F.; Wan, X.; Lu, W.; Xie, X.; Cheng, X. miR-375 mediated acquired chemo-resistance in cervical cancer by facilitating EMT. PLoS ONE 2014, 9, e109299.
  112. Babion, I.; Miok, V.; Jaspers, A.; Huseinovic, A.; Steenbergen, R.D.M.; van Wieringen, W.N.; Wilting, S.M. Identification of Deregulated Pathways.; Key Regulators.; and Novel miRNA-mRNA Interactions in HPV-Mediated Transformation. Cancers 2020, 12, 700.
  113. Song, L.; Liu, S.; Zhang, L.; Yao, H.; Gao, F.; Xu, D.; Li, Q. MiR-21 modulates radiosensitivity of cervical cancer through inhibiting autophagy via the PTEN/Akt/HIF-1α feedback loop and the Akt-mTOR signaling pathway. Tumour Biol. 2016, 37, 12161–12168.
  114. Chen, R.; Gan, Q.; Zhao, S.; Zhang, D.; Wang, S.; Yao, L.; Yuan, M.; Cheng, J. DNA methylation of miR-138 regulates cell proliferation and EMT in cervical cancer by targeting EZH2. BMC Cancer 2022, 22, 488.
  115. Zheng, Q.; Peskoe, S.B.; Ribas, J.; Rafiqi, F.; Kudrolli, T.; Meeker, A.K.; De Marzo, A.M.; Platz, E.A.; Lupold, S.E. Investigation of miR-21, miR-141, and miR-221 expression levels in prostate adenocarcinoma for associated risk of recurrence after radical prostatectomy. Prostate 2014, 74, 1655–1662.
  116. Xu, S.; Ge, J.; Zhang, Z.; Zhou, W. miR-141 inhibits prostatic cancer cell proliferation and migration, and induces cell apoptosis via targeting of RUNX1. Oncol. Rep. 2018, 39, 1454–1460.
  117. Hirata, H.; Ueno, K.; Shahryari, V.; Deng, G.; Tanaka, Y.; Tabatabai, Z.L.; Hinoda, Y.; Dahiya, R. MicroRNA-182-5p promotes cell invasion and proliferation by down regulating FOXF2.; RECK and MTSS1 genes in human prostate cancer. PLoS ONE 2013, 8, e55502.
  118. Peng, X.; Guo, W.; Liu, T.; Wang, X.; Tu, X.; Xiong, D.; Chen, S.; Lai, Y.; Du, H.; Chen, G.; et al. Identification of miRs-143 and -145 that is associated with bone metastasis of prostate cancer and involved in the regulation of EMT. PLoS ONE 2011, 6, e20341.
  119. Liu, J.; Li, J.; Ma, Y.; Xu, C.; Wang, Y.; He, Y. MicroRNA miR-145-5p inhibits Phospholipase D 5 (PLD5) to downregulate cell proliferation and metastasis to mitigate prostate cancer. Bioengineered 2021, 12, 3240–3251.
  120. Bhatnagar, N.; Li, X.; Padi, S.K.; Zhang, Q.; Tang, M.S.; Guo, B. Downregulation of miR-205 and miR-31 confers resistance to chemotherapy-induced apoptosis in prostate cancer cells. Cell Death Dis. 2010, 1, e105.
  121. Galardi, S.; Mercatelli, N.; Giorda, E.; Massalini, S.; Frajese, G.V.; Ciafrè, S.A.; Farace, M.G. miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J. Biol. Chem. 2007, 282, 23716–23724.
  122. Schubert, M.; Spahn, M.; Kneitz, S.; Scholz, C.J.; Joniau, S.; Stroebel, P.; Riedmiller, H.; Kneitz, B. Distinct microRNA expression profile in prostate cancer patients with early clinical failure and the impact of let-7 as prognostic marker in high-risk prostate cancer. PLoS ONE 2013, 8, e65064.
  123. Li, T.; Li, D.; Sha, J.; Sun, P.; Huang, Y. MicroRNA-21 directly targets MARCKS and promotes apoptosis resistance and invasion in prostate cancer cells. Biochem. Biophys. Res. Commun. 2009, 383, 280–285.
  124. Liu, L.Z.; Li, C.; Chen, Q.; Jing, Y.; Carpenter, R.; Jiang, Y.; Kung, H.F.; Lai, L.; Jiang, B.H. MiR-21 induced angiogenesis through AKT and ERK activation and HIF-1α expression. PLoS ONE 2011, 6, e19139.
  125. Palmer, R.D.; Murray, M.J.; Saini, H.K.; van Dongen, S.; Abreu-Goodger, C.; Muralidhar, B.; Pett, M.R.; Thornton, C.M.; Nicholson, J.C.; Enright, A.J.; et al. Malignant germ cell tumors display common microRNA profiles resulting in global changes in expression of messenger RNA targets. Cancer Res. 2010, 70, 2911–2923.
  126. Looijenga, L.H.; Gillis, A.J.; Stoop, H.J.; Hersmus, R.; Oosterhuis, J.W. Chromosomes and expression in human testicular germ-cell tumors: Insight into their cell of origin and pathogenesis. Ann. N. Y. Acad. Sci. 2007, 1120, 187–214.
  127. Voorhoeve, P.M.; le Sage, C.; Schrier, M.; Gillis, A.J.; Stoop, H.; Nagel, R.; Liu, Y.P.; van Duijse, J.; Drost, J.; Griekspoor, A.; et al. A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell 2006, 124, 1169–1181.
  128. Liu, J.; Shi, H.; Li, X.; Chen, G.; Larsson, C.; Lui, W.O. miR-223-3p regulates cell growth and apoptosis via FBXW7 suggesting an oncogenic role in human testicular germ cell tumors. Int. J. Oncol. 2017, 50, 356–364.
  129. Huang, H.; Tian, H.; Duan, Z.; Cao, Y.; Zhang, X.S.; Sun, F. microRNA-383 impairs phosphorylation of H2AX by targeting PNUTS and inducing cell cycle arrest in testicular embryonal carcinoma cells. Cell Signal. 2014, 26, 903–911.
  130. Das, M.K.; Evensen, H.S.F.; Furu, K.; Haugen, T.B. miRNA-302s may act as oncogenes in human testicular germ cell tumours. Sci. Rep. 2019, 9, 9189.
  131. Liu, L.; Lian, J.; Zhang, H.; Tian, H.; Liang, M.; Yin, M.; Sun, F. MicroRNA-302a sensitizes testicular embryonal carcinoma cells to cisplatin-induced cell death. J. Cell. Physiol. 2013, 228, 2294–2304.
  132. Özata, D.M.; Li, X.; Lee, L.; Liu, J.; Warsito, D.; Hajeri, P.; Hultman, I.; Fotouhi, O.; Marklund, S.; Ährlund-Richter, L.; et al. Loss of miR-514a-3p regulation of PEG3 activates the NF-kappa B pathway in human testicular germ cell tumors. Cell Death Dis. 2017, 8, e2759.
  133. Chen, B.F.; Gu, S.; Suen, Y.K.; Li, L.; Chan, W.Y. microRNA-199a-3p, DNMT3A, and aberrant DNA methylation in testicular cancer. Epigenetics 2014, 9, 119–128.
  134. Zhao, G.; Yin, Y.; Zhao, B. miR-140-5p is negatively correlated with proliferation.; invasion.; and tumorigenesis in malignant melanoma by targeting SOX4 via the Wnt/β-catenin and NF-κB cascades. J. Cell. Physiol. 2020, 235, 2161–2170.
  135. Bemis, L.T.; Chen, R.; Amato, C.M.; Classen, E.H.; Robinson, S.E.; Coffey, D.G.; Erickson, P.F.; Shellman, Y.G.; Robinson, W.A. MicroRNA-137 targets microphthalmia-associated transcription factor in melanoma cell lines. Cancer Res. 2008, 68, 1362–1368.
  136. Yan, D.; Dong, X.D.; Chen, X.; Yao, S.; Wang, L.; Wang, J.; Wang, C.; Hu, D.N.; Qu, J.; Tu, L. Role of microRNA-182 in posterior uveal melanoma: Regulation of tumor development through MITF, BCL2 and cyclin D2. PLoS ONE 2012, 7, e40967.
  137. Haflidadóttir, B.S.; Bergsteinsdóttir, K.; Praetorius, C.; Steingrímsson, E. miR-148 regulates Mitf in melanoma cells. PLoS ONE 2010, 5, e11574.
  138. Margue, C.; Philippidou, D.; Reinsbach, S.E.; Schmitt, M.; Behrmann, I.; Kreis, S. New target genes of MITF-induced microRNA-211 contribute to melanoma cell invasion. PLoS ONE 2013, 8, e73473.
  139. Sun, X.; Li, J.; Sun, Y.; Zhang, Y.; Dong, L.; Shen, C.; Yang, L.; Yang, M.; Li, Y.; Shen, G.; et al. miR-7 reverses the resistance to BRAFi in melanoma by targeting EGFR/IGF-1R/CRAF and inhibiting the MAPK and PI3K/AKT signaling pathways. Oncotarget 2016, 7, 53558–53570.
  140. Rambow, F.; Job, B.; Petit, V.; Gesbert, F.; Delmas, V.; Seberg, H.; Meurice, G.; Van Otterloo, E.; Dessen, P.; Robert, C.; et al. New Functional Signatures for Understanding Melanoma Biology from Tumor Cell Lineage-Specific Analysis. Cell Rep. 2015, 13, 840–853.
  141. Arts, N.; Cané, S.; Hennequart, M.; Lamy, J.; Bommer, G.; Van den Eynde, B.; De Plaen, E. microRNA-155, induced by interleukin-1ß, Represses the expression of microphthalmia-associated transcription factor (MITF-M) in melanoma cells. PLoS ONE 2015, 10, e0122517.
  142. Philippidou, D.; Schmitt, M.; Moser, D.; Margue, C.; Nazarov, P.V.; Muller, A.; Vallar, L.; Nashan, D.; Behrmann, I.; Kreis, S. Signatures of microRNAs and selected microRNA target genes in human melanoma. Cancer Res. 2010, 70, 4163–4173.
  143. Xu, N.; Zhang, L.; Meisgen, F.; Harada, M.; Heilborn, J.; Homey, B.; Grandér, D.; Ståhle, M.; Sonkoly, E.; Pivarcsi, A. MicroRNA-125b down-regulates matrix metallopeptidase 13 and inhibits cutaneous squamous cell carcinoma cell proliferation, migration, and invasion. J. Biol. Chem. 2012, 287, 29899–29908.
  144. Toll, A.; Salgado, R.; Espinet, B.; Díaz-Lagares, A.; Hernández-Ruiz, E.; Andrades, E.; Sandoval, J.; Esteller, M.; Pujol, R.M.; Hernández-Muñoz, I. MiR-204 silencing in intraepithelial to invasive cutaneous squamous cell carcinoma progression. Mol. Cancer 2016, 15, 53.
  145. Neu, J.; Dziunycz, P.J.; Dzung, A.; Lefort, K.; Falke, M.; Denzler, R.; Freiberger, S.N.; Iotzova-Weiss, G.; Kuzmanov, A.; Levesque, M.P.; et al. miR-181a decelerates proliferation in cutaneous squamous cell carcinoma by targeting the proto-oncogene KRAS. PLoS ONE 2017, 12, e0185028.
  146. Bai, X.; Zhou, Y.; Chen, P.; Yang, M.; Xu, J. MicroRNA-142-5p induces cancer stem cell-like properties of cutaneous squamous cell carcinoma via inhibiting PTEN. J. Cell. Biochem. 2018, 119, 2179–2188.
  147. Gong, Z.H.; Zhou, F.; Shi, C.; Xiang, T.; Zhou, C.K.; Wang, Q.Q.; Jiang, Y.S.; Gao, S.F. miRNA-221 promotes cutaneous squamous cell carcinoma progression by targeting PTEN. Cell. Mol. Biol. Lett. 2019, 24, 9.
  148. Lohcharoenkal, W.; Harada, M.; Lovén, J.; Meisgen, F.; Landén, N.X.; Zhang, L.; Lapins, J.; Mahapatra, K.D.; Shi, H.; Nissinen, L.; et al. MicroRNA-203 Inversely Correlates with Differentiation Grade, Targets c-MYC, and Functions as a Tumor Suppressor in cSCC. J. Investig. Dermatol. 2016, 136, 2485–2494.
  149. Luo, Q.; Li, W.; Zhao, T.; Tian, X.; Liu, Y.; Zhang, X. Role of miR-148a in cutaneous squamous cell carcinoma by repression of MAPK pathway. Arch. Biochem. Biophys. 2015, 583, 47–54.
  150. Wang, S.H.; Zhou, J.D.; He, Q.Y.; Yin, Z.Q.; Cao, K.; Luo, C.Q. MiR-199a inhibits the ability of proliferation and migration by regulating CD44-Ezrin signaling in cutaneous squamous cell carcinoma cells. Int. J. Clin. Exp. Pathol. 2014, 7, 7131–7141.
  151. Yamane, K.; Jinnin, M.; Etoh, T.; Kobayashi, Y.; Shimozono, N.; Fukushima, S.; Masuguchi, S.; Maruo, K.; Inoue, Y.; Ishihara, T.; et al. Down-regulation of miR-124/-214 in cutaneous squamous cell carcinoma mediates abnormal cell proliferation via the induction of ERK. J. Mol. Med. 2013, 91, 69–81.
  152. Pallante, P.; Visone, R.; Ferracin, M.; Ferraro, A.; Berlingieri, M.T.; Troncone, G.; Chiappetta, G.; Liu, C.G.; Santoro, M.; Negrini, M.; et al. MicroRNA deregulation in human thyroid papillary carcinomas. Endocr. Relat. Cancer 2006, 13, 497–508.
  153. He, H.; Jazdzewski, K.; Li, W.; Liyanarachchi, S.; Nagy, R.; Volinia, S.; Calin, G.A.; Liu, C.G.; Franssila, K.; Suster, S.; et al. The role of microRNA genes in papillary thyroid carcinoma. Proc. Natl. Acad. Sci. USA 2005, 102, 19075–19080.
  154. Visone, R.; Russo, L.; Pallante, P.; De Martino, I.; Ferraro, A.; Leone, V.; Borbone, E.; Petrocca, F.; Alder, H.; Croce, C.M.; et al. MicroRNAs (miR)-221 and miR-222, both overexpressed in human thyroid papillary carcinomas, regulate p27Kip1 protein levels and cell cycle. Endocr. Relat. Cancer 2007, 14, 791–798.
  155. Nikiforova, M.N.; Tseng, G.C.; Steward, D.; Diorio, D.; Nikiforov, Y.E. MicroRNA expression profiling of thyroid tumors: Biological significance and diagnostic utility. J. Clin. Endocrinol. Metab. 2008, 93, 1600–1608.
  156. Liao, B.; Liu, S.; Liu, J.; Reddy, P.A.K.; Ying, Y.; Xie, Y.; Wang, J.; Zeng, X. Long Noncoding RNA CTC Inhibits Proliferation and Invasion by Targeting miR-146 to Regulate KIT in Papillary Thyroid Carcinoma. Sci. Rep. 2020, 10, 4616.
  157. Czajka, A.A.; Wójcicka, A.; Kubiak, A.; Kotlarek, M.; Bakuła-Zalewska, E.; Koperski, Ł.; Wiechno, W.; Jażdżewski, K. Family of microRNA-146 Regulates RARβ in Papillary Thyroid Carcinoma. PLoS ONE 2016, 11, e0151968.
  158. Braun, J.; Hoang-Vu, C.; Dralle, H.; Hüttelmaier, S. Downregulation of microRNAs directs the EMT and invasive potential of anaplastic thyroid carcinomas. Oncogene 2010, 29, 4237–4244.
  159. Zhang, Z.; Liu, Z.B.; Ren, W.M.; Ye, X.G.; Zhang, Y.Y. The miR-200 family regulates the epithelial-mesenchymal transition induced by EGF/EGFR in anaplastic thyroid cancer cells. Int. J. Mol. Med. 2012, 30, 856–862.
  160. Chen, G.; Gao, Y.; Wang, G.; Dai, G.; Tong, L. MiR-145 inhibits the migration and invasion of papillary thyroid carcinoma cells through NF-κB pathway regulation. J. Cell. Biochem. 2020, 121, 3325–3332.
  161. Hudson, J.; Duncavage, E.; Tamburrino, A.; Salerno, P.; Xi, L.; Raffeld, M.; Moley, J.; Chernock, R.D. Overexpression of miR-10a and miR-375 and downregulation of YAP1 in medullary thyroid carcinoma. Exp. Mol. Pathol. 2013, 95, 62–67.
  162. Abraham, D.; Jackson, N.; Gundara, J.S.; Zhao, J.; Gill, A.J.; Delbridge, L.; Robinson, B.G.; Sidhu, S.B. MicroRNA profiling of sporadic and hereditary medullary thyroid cancer identifies predictors of nodal metastasis, prognosis, and potential therapeutic targets. Clin. Cancer Res. 2011, 17, 4772–4781.
  163. Pennelli, G.; Galuppini, F.; Barollo, S.; Cavedon, E.; Bertazza, L.; Fassan, M.; Guzzardo, V.; Pelizzo, M.R.; Rugge, M.; Mian, C. The PDCD4/miR-21 pathway in medullary thyroid carcinoma. Hum. Pathol. 2015, 46, 50–57.
  164. Gao, Y.; Luo, L.H.; Li, S.; Yang, C. miR-17 inhibitor suppressed osteosarcoma tumor growth and metastasis via increasing PTEN expression. Biochem. Biophys. Res. Commun. 2014, 444, 230–234.
  165. McKinsey, E.L.; Parrish, J.K.; Irwin, A.E.; Niemeyer, B.F.; Kern, H.B.; Birks, D.K.; Jedlicka, P. A novel oncogenic mechanism in Ewing sarcoma involving IGF pathway targeting by EWS/Fli1-regulated microRNAs. Oncogene 2011, 30, 4910–4920.
  166. Wang, Y.; Zhang, S.; Xu, Y.; Zhang, Y.; Guan, H.; Li, X.; Li, Y.; Wang, Y. Upregulation of miR-192 inhibits cell growth and invasion and induces cell apoptosis by targeting TCF7 in human osteosarcoma. Tumour Biol. 2016, 37, 15211–15220.
  167. Gindin, Y.; Jiang, Y.; Francis, P.; Walker, R.L.; Abaan, O.D.; Zhu, Y.J.; Meltzer, P.S. miR-23a impairs bone differentiation in osteosarcoma via down-regulation of GJA1. Front. Genet. 2015, 6, 233.
  168. He, Y.; Meng, C.; Shao, Z.; Wang, H.; Yang, S. MiR-23a functions as a tumor suppressor in osteosarcoma. Cell. Physiol. Biochem. 2014, 34, 1485–1496.
  169. Llobat, L.; Gourbault, O. Role of MicroRNAs in Human Osteosarcoma: Future Perspectives. Biomedicines 2021, 9, 463.
  170. Sharma, P.C.; Gupta, A. MicroRNAs: Potential biomarkers for diagnosis and prognosis of different cancers. Transl. Cancer Res. 2020, 9, 5798–5818.
  171. Musilova, K.; Mraz, M. MicroRNAs in B-cell lymphomas: How a complex biology gets more complex. Leukemia 2015, 29, 1004–1017.
  172. Olive, V.; Li, Q.; He, L. mir-17-92: A polycistronic oncomir with pleiotropic functions. Immunol. Rev. 2013, 253, 158–166.
  173. O’Donnell, K.A.; Wentzel, E.A.; Zeller, K.I.; Dang, C.V.; Mendell, J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005, 435, 839–843.
  174. Korać, P.; Antica, M.; Matulić, M. MiR-7 in Cancer Development. Biomedicines 2021, 9, 325.
  175. Masoudi, M.S.; Mehrabian, E.; Mirzaei, H. MiR-21: A key player in glioblastoma pathogenesis. J. Cell. Biochem. 2018, 119, 1285–1290.
  176. Hamamoto, J.; Soejima, K.; Yoda, S.; Naoki, K.; Nakayama, S.; Satomi, R.; Terai, H.; Ikemura, S.; Sato, T.; Yasuda, H.; et al. Identification of microRNAs differentially expressed between lung squamous cell carcinoma and lung adenocarcinoma. Mol. Med. Rep. 2013, 8, 456–462.
  177. Landi, M.T.; Zhao, Y.; Rotunno, M.; Koshiol, J.; Liu, H.; Bergen, A.W.; Rubagotti, M.; Goldstein, A.M.; Linnoila, I.; Marincola, F.M.; et al. MicroRNA expression differentiates histology and predicts survival of lung cancer. Clin. Cancer Res. 2010, 16, 430–441.
  178. Lebanony, D.; Benjamin, H.; Gilad, S.; Ezagouri, M.; Dov, A.; Ashkenazi, K.; Gefen, N.; Izraeli, S.; Rechavi, G.; Pass, H.; et al. Diagnostic assay based on hsa-miR-205 expression distinguishes squamous from nonsquamous non-small-cell lung carcinoma. J. Clin. Oncol. 2009, 27, 2030–2037.
  179. Bishop, J.A.; Benjamin, H.; Cholakh, H.; Chajut, A.; Clark, D.P.; Westra, W.H. Accurate classification of non-small cell lung carcinoma using a novel microRNA-based approach. Clin. Cancer Res. 2010, 16, 610–619.
  180. El Founini, Y.; Chaoui, I.; Dehbi, H.; El Mzibri, M.; Abounader, R.; Guessous, F. MicroRNAs: Key Regulators in Lung Cancer. Microrna 2021, 10, 109–122.
  181. Fang, R.; Xiao, T.; Fang, Z.; Sun, Y.; Li, F.; Gao, Y.; Feng, Y.; Li, L.; Wang, Y.; Liu, X.; et al. MicroRNA-143 (miR-143) regulates cancer glycolysis via targeting hexokinase 2 gene. J. Biol. Chem. 2012, 287, 23227–23235.
  182. Wang, G.; Mao, W.; Zheng, S. MicroRNA-183 regulates Ezrin expression in lung cancer cells. FEBS Lett. 2008, 582, 3663–3668.
  183. Xie, X.; Tan, W.; Chen, B.; Huang, X.; Peng, C.; Yan, S.; Yang, L.; Song, C.; Wang, J.; Zheng, W.; et al. Preoperative prediction nomogram based on primary tumor miRNAs signature and clinical-related features for axillary lymph node metastasis in early-stage invasive breast cancer. Int. J. Cancer 2018, 142, 1901–1910.
  184. Quesne, J.L.; Jones, J.; Warren, J.; Dawson, S.J.; Ali, H.R.; Bardwell, H.; Blows, F.; Pharoah, P.; Caldas, C. Biological and prognostic associations of miR-205 and let-7b in breast cancer revealed by in situ hybridization analysis of micro-RNA expression in arrays of archival tumour tissue. J. Pathol. 2012, 227, 306–314.
  185. Kalinkova, L.; Nikolaieva, N.; Smolkova, B.; Ciernikova, S.; Kajo, K.; Bella, V.; Kajabova, V.H.; Kosnacova, H.; Minarik, G.; Fridrichova, I. miR-205-5p Downregulation and ZEB1 Upregulation Characterize the Disseminated Tumor Cells in Patients with Invasive Ductal Breast Cancer. Int. J. Mol. Sci. 2021, 23, 103.
  186. Abolghasemi, M.; Tehrani, S.S.; Yousefi, T.; Karimian, A.; Mahmoodpoor, A.; Ghamari, A.; Jadidi-Niaragh, F.; Yousefi, M.; Kafil, H.S.; Bastami, M.; et al. MicroRNAs in breast cancer: Roles, functions, and mechanism of actions. J. Cell. Physiol. 2020, 235, 5008–5029.
  187. Ghafouri-Fard, S.; Shirvani-Farsani, Z.; Branicki, W.; Taheri, M. MicroRNA Signature in Renal Cell Carcinoma. Front. Oncol. 2020, 10, 596359.
  188. Chen, X.; Wang, L.; Qu, J.; Guan, N.N.; Li, J.Q. Predicting miRNA–disease association based on inductive matrix completion. Bioinformatics 2018, 34, 4256–4265.
  189. Khashkhashi Moghadam, S.; Bakhshinejad, B.; Khalafizadeh, A.; Mahmud Hussen, B.; Babashah, S. Non-coding RNA-associated competitive endogenous RNA regulatory networks: Novel diagnostic and therapeutic opportunities for hepatocellular carcinoma. J. Cell. Mol. Med. 2022, 26, 287–305.
  190. Liu, L.; Wang, Y.; Bai, R.; Yang, K.; Tian, Z. MiR-186 inhibited aerobic glycolysis in gastric cancer via HIF-1α regulation. Oncogenesis 2016, 5, e224.
  191. Yue, Y.; Lin, X.; Qiu, X.; Yang, L.; Wang, R. The Molecular Roles and Clinical Implications of Non-Coding RNAs in Gastric Cancer. Front. Cell Dev. Biol. 2021, 9, 802745.
  192. Bañuelos-Villegas, E.G.; Pérez-y Pérez, M.F.; Alvarez-Salas, L.M. Cervical Cancer. Papillomavirus. and miRNA Dysfunction. Front. Mol. Biosci. 2021, 8, 758337.
  193. Gillis, A.J.; Stoop, H.J.; Hersmus, R.; Oosterhuis, J.W.; Sun, Y.; Chen, C.; Guenther, S.; Sherlock, J.; Veltman, I.; Baeten, J.; et al. High-throughput microRNAome analysis in human germ cell tumours. J. Pathol. 2007, 213, 319–328.
  194. Das, M.K.; Haugen, Ø.P.; Haugen, T.B. Diverse Roles and Targets of miRNA in the Pathogenesis of Testicular Germ Cell Tumour. Cancers 2022, 14, 1190.
  195. Varrone, F.; Caputo, E. The miRNAs Role in Melanoma and in Its Resistance to Therapy. Int. J. Mol. Sci. 2020, 21, 878.
  196. Simmons, J.L.; Pierce, C.J.; Al-Ejeh, F.; Boyle, G.M. MITF and BRN2 contribute to metastatic growth after dissemination of melanoma. Sci. Rep. 2017, 7, 10909.
  197. Fuziwara, C.S.; Kimura, E.T. MicroRNA Deregulation in Anaplastic Thyroid Cancer Biology. Int. J. Endocrinol. 2014, 2014, 743450.
More
This entry is offline, you can click here to edit this entry!
Video Production Service