In vitro and in vivo anti-tumor activity of miR-221/222 inhibitors in multiple myeloma

Maria Teresa Di Martino, Annamaria Gullà, Maria Eugenia Gallo Cantafio, Marta Lionetti, Emanuela Leone, Nicola Amodio, Pietro Hiram Guzzi, Umberto Foresta, Francesco Conforti, Mario Cannataro, Antonino Neri, Antonio Giordano, Pierosandro Tagliaferri, Pierfrancesco Tassone, Maria Teresa Di Martino, Annamaria Gullà, Maria Eugenia Gallo Cantafio, Marta Lionetti, Emanuela Leone, Nicola Amodio, Pietro Hiram Guzzi, Umberto Foresta, Francesco Conforti, Mario Cannataro, Antonino Neri, Antonio Giordano, Pierosandro Tagliaferri, Pierfrancesco Tassone

Abstract

A rising body of evidence suggests that silencing microRNAs (miRNAs) with oncogenic potential may represent a successful therapeutic strategy for human cancer. We investigated the therapeutic activity of miR-221/222 inhibitors against human multiple myeloma (MM) cells. Enforced expression of miR-221/222 inhibitors triggered in vitro anti-proliferative effects and up-regulation of canonic miR-221/222 targets, including p27Kip1, PUMA, PTEN and p57Kip2, in MM cells highly expressing miR-221/222. Conversely, transfection of miR-221/222 mimics increased S-phase and down-regulated p27Kip1 protein expression in MM with low basal miR-221/222 levels. The effects of miR-221/222 inhibitors was also evaluated in MM xenografts in SCID/ NOD mice. Significant anti-tumor activity was achieved in xenografted mice by the treatment with miR-221/222 inhibitors, together with up-regulation of canonic protein targets in tumors retrieved from animals. These findings provide proof of principle that silencing the miR-221/222 cluster exerts significant therapeutic activity in MM cells with high miR-221/222 level of expression, which mostly occurs in TC2 and TC4 MM groups. These findings suggest that MM genotyping may predict the therapeutic response. All together our results support a framework for clinical development of miR-221/222 inhibitors-based therapeutic strategy in this still incurable disease.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. miR-221 and miR-222 expression in…
Figure 1. miR-221 and miR-222 expression in primary CD138+ normal plasma cells, primary MM and PCL cells and established MM cell lines
A) Differential expression of miR-221 and miR-222 in immunoselected CD138+ cells from 3 healthy donors, 38 MM and 2 PCL, by microarray analysis. Expression values were normalized by aroma.light-package for Bioconductor. MM were TC classified according to the presence of the recurrent IGH chromosomal translocations and cyclins D expression as previously described (30). miR-221 and miR-222 are reported as raw expression values. Statistical significance was assessed by SAM multi-class analysis, (q-value=0). N(1-3): CD138+ cells from normal healthy donors. MM and PCL were numbered referring to individual patients in the original data set. B) Differential expression of miR-221 and miR-222 in 16 MM cell lines by Affymetrix GeneChip® miRNA 1.0 Array. Histogram bars indicate miR-221 or miR-222 expression values normalized by miRNA QC Tool (Affymetrix).
Figure 2. Biological effects induced by transient…
Figure 2. Biological effects induced by transient expression of miR-221/222 in MM cell lines
A) Cell cycle perturbation in U266 cells induced by transient pre-miR221/222 enforced expression. At least 20,000 events for each point were analyzed in 3 independent experiments. Results are representative of one out of 3 experiments 72 hours after transfection. B) Effects induced on BrdU uptake of RPMI-8226 cells by transfection with pre-miR-221/222 mimics or scrambled sequences. Averaged values ±SD from 3 independent experiments are plotted. C) p27Kip1 protein expression 48-72 and 96 hours after transfection of U266 and RPMI-8226 cells with pre-miR-221/222 mimics or scrambled controls (NC) was assessed by Western blotting assay. D) Inverse correlation between p27Kip1 mRNA and miR-221 levels in a dataset of 40 (38 MM + 2 PCL) patients is shown. The red bars represent mature miR-221 expression (vertical axis on right side) and the spots represent target gene (p27Kip1 mRNA) expression (vertical axis on left side). Horizontal axis: patient samples ordered according to increasing p27Kip1 expression.
Figure 3. In vitro anti-proliferative activity of…
Figure 3. In vitro anti-proliferative activity of miR-221 and miR-222 inhibitors on MM cell lines
A-B) Cell growth analysis of OPM2 (A) and NCI-H929 (B) cells transfected with miR-221/222 inhibitors or scrambled oligonucleotides (NC). Analysis was performed by trypan blue exclusion assay. Averaged values of 3 independent experiments are plotted in each frame including +SD. Significant P values (P

Figure 4. Molecular effects induced by miR-221/222…

Figure 4. Molecular effects induced by miR-221/222 inhibitors in MM cells

A) miR-221 and miR-222…

Figure 4. Molecular effects induced by miR-221/222 inhibitors in MM cells
A) miR-221 and miR-222 q-RT-PCR 24 hours after transfection with miR-221/222 inhibitors and NC in OPM2 cells. The results are shown as miRNA expression level after normalization with RNU44 and ΔΔCt calculations. Data represent the average of 3 independent experiments +SD. B) q-RT-PCR of p27Kip1, PUMA, PTEN and p57Kip2 mRNA expression 24 hours after transfection with miR-221/222 inhibitors or NC in OPM2 cells. The results are shown as average mRNA expression after normalization with GAPDH and ΔΔCt calculations. Data represent the average of 3 independent experiments +SD. (*) P

Figure 5. Whole Gene Expression Profiles of…

Figure 5. Whole Gene Expression Profiles of MM cells after miR-221/222 knockdown

A) Hierarchical clustering…

Figure 5. Whole Gene Expression Profiles of MM cells after miR-221/222 knockdown
A) Hierarchical clustering of differentially expressed genes between miR-221/222 inhibitors or NC treated OPM2 cells by Gene 1.0 ST array chip (Affymetrix) and DChip software. Genes that underwent a 0.5-fold change as compared to control, were selected and clustered. Assays performed in triplicate are shown. B) Pathways modulated by miR-221/222 inhibitors in OPM2 MM cells are shown. Analysis was performed by Ingenuity Pathway Analysis on genes significantly modulated (FC>0.5). The bar graphs show pathways most modulated by miR-221/222 inhibitors as compared to control, based on statistical significance (P-value and ratio). The yellow line indicates the threshold of significance. C) Functional network scenario with principal nodes in the most perturbed pathways are showed. Up- and down-regulated genes are indicated in red and green, respectively. Continuous and discontinuous lines represent direct and indirect functional and physical interactions between genes as previously reported.

Figure 6. In vivo anti-tumor activity of…

Figure 6. In vivo anti-tumor activity of miR-221/222 inhibitors in MM xenografted SCID/NOD mice

A)…

Figure 6. In vivo anti-tumor activity of miR-221/222 inhibitors in MM xenografted SCID/NOD mice
A) Effects of formulated or unformulated miR-221/222 inhibitors. Subcutaneous OPM2 xenografts were treated every 2 days with 20 μg of formulated(NLE)-miR-221/222 inhibitors or unformulated-miR221/222 inhibitors. As control 2 separate groups of tumor-bearing animals were injected with formulated(NLE)-NC or unformulated-NC. B) Effects of treatments with formulated miR-221 or miR-222 individual inhibitors. Subcutaneous OPM2 xenografts were repeatedly treated every 2 days, with 20 μg of NLE-miR-221 inhibitors, or NLE-miR-222 inhibitors, or NLE-NC. C) Effects of unformulated-miR-221 inhibitors or NC. Subcutaneous OPM2 xenografts were repeatedly treated every 2 days with 20 μg of unformulated-miR-221 inhibitors, or unformulated-NC. The tumor volumes averages of each group +SD are reported. In each experiment P values were calculated for miRNA inhibitors versus scrambled oligonucleotides (NC) and significant P values (P<0.05) are indicated by stars.

Figure 7. miR-221 activity and targets silencing…

Figure 7. miR-221 activity and targets silencing in retrieved MM xenografted tumors

A) q-RT-PCR of…

Figure 7. miR-221 activity and targets silencing in retrieved MM xenografted tumors
A) q-RT-PCR of miR-221 and miR-222 in retrieved tumors treated with unformulated-miR-221 inhibitors or unformulated-NC. The results are shown as averaged miRNA expression after normalization with RNU44 and ΔΔCt calculations as compared to control (miRNA expression in NC treated animals). B) H&E (20-fold magnification) and immunohistochemistry (40-fold magnification) staining of xenografted tumors retrieved from treated animals. C) q-RT-PCR of p27Kip1 and PTEN mRNA levels in treated tumors retrieved from mice. Raw Ct values were normalized to GAPDH housekeeping mRNA and expressed as ΔΔCt values calculated using the comparative cross threshold method. D) Western blot analysis of p27Kip1 and PTEN protein in retrieved tumors from mice treated with miR-221 inhibitors or NC. D-E) Western blot analysis of total AKT and p-AKT (D) and total ERK and p-ERK (E) in retrieved tumors from mice treated with miR-221 inhibitors or NC. Representative experiments are shown in figure. γ-tubulin was the protein loading control.
All figures (7)
Similar articles
Cited by
References
    1. Anderson KC, Carrasco RD. Pathogenesis of myeloma. Annu Rev Pathol. 2011;6:249–274. - PubMed
    1. Rajkumar SV. Treatment of multiple myeloma. Nat Rev Clin Oncol. 2011;8(8):479–491. - PMC - PubMed
    1. Morabito F, Recchia AG, Mazzone C, Gentile M. Targeted therapy of multiple myeloma: the changing paradigm at the beginning of the new millennium. Current cancer drug targets. 2012;12(7):743–756. - PubMed
    1. Lonial S, Mitsiades CS, Richardson PG. Treatment options for relapsed and refractory multiple myeloma. Clin Cancer Res. 2011;17(6):1264–1277. - PubMed
    1. Kapoor P, Rajkumar SV. Update on risk stratification and treatment of newly diagnosed multiple myeloma. Int J Hematol. 2011 - PubMed
Show all 59 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4. Molecular effects induced by miR-221/222…
Figure 4. Molecular effects induced by miR-221/222 inhibitors in MM cells
A) miR-221 and miR-222 q-RT-PCR 24 hours after transfection with miR-221/222 inhibitors and NC in OPM2 cells. The results are shown as miRNA expression level after normalization with RNU44 and ΔΔCt calculations. Data represent the average of 3 independent experiments +SD. B) q-RT-PCR of p27Kip1, PUMA, PTEN and p57Kip2 mRNA expression 24 hours after transfection with miR-221/222 inhibitors or NC in OPM2 cells. The results are shown as average mRNA expression after normalization with GAPDH and ΔΔCt calculations. Data represent the average of 3 independent experiments +SD. (*) P

Figure 5. Whole Gene Expression Profiles of…

Figure 5. Whole Gene Expression Profiles of MM cells after miR-221/222 knockdown

A) Hierarchical clustering…

Figure 5. Whole Gene Expression Profiles of MM cells after miR-221/222 knockdown
A) Hierarchical clustering of differentially expressed genes between miR-221/222 inhibitors or NC treated OPM2 cells by Gene 1.0 ST array chip (Affymetrix) and DChip software. Genes that underwent a 0.5-fold change as compared to control, were selected and clustered. Assays performed in triplicate are shown. B) Pathways modulated by miR-221/222 inhibitors in OPM2 MM cells are shown. Analysis was performed by Ingenuity Pathway Analysis on genes significantly modulated (FC>0.5). The bar graphs show pathways most modulated by miR-221/222 inhibitors as compared to control, based on statistical significance (P-value and ratio). The yellow line indicates the threshold of significance. C) Functional network scenario with principal nodes in the most perturbed pathways are showed. Up- and down-regulated genes are indicated in red and green, respectively. Continuous and discontinuous lines represent direct and indirect functional and physical interactions between genes as previously reported.

Figure 6. In vivo anti-tumor activity of…

Figure 6. In vivo anti-tumor activity of miR-221/222 inhibitors in MM xenografted SCID/NOD mice

A)…

Figure 6. In vivo anti-tumor activity of miR-221/222 inhibitors in MM xenografted SCID/NOD mice
A) Effects of formulated or unformulated miR-221/222 inhibitors. Subcutaneous OPM2 xenografts were treated every 2 days with 20 μg of formulated(NLE)-miR-221/222 inhibitors or unformulated-miR221/222 inhibitors. As control 2 separate groups of tumor-bearing animals were injected with formulated(NLE)-NC or unformulated-NC. B) Effects of treatments with formulated miR-221 or miR-222 individual inhibitors. Subcutaneous OPM2 xenografts were repeatedly treated every 2 days, with 20 μg of NLE-miR-221 inhibitors, or NLE-miR-222 inhibitors, or NLE-NC. C) Effects of unformulated-miR-221 inhibitors or NC. Subcutaneous OPM2 xenografts were repeatedly treated every 2 days with 20 μg of unformulated-miR-221 inhibitors, or unformulated-NC. The tumor volumes averages of each group +SD are reported. In each experiment P values were calculated for miRNA inhibitors versus scrambled oligonucleotides (NC) and significant P values (P<0.05) are indicated by stars.

Figure 7. miR-221 activity and targets silencing…

Figure 7. miR-221 activity and targets silencing in retrieved MM xenografted tumors

A) q-RT-PCR of…

Figure 7. miR-221 activity and targets silencing in retrieved MM xenografted tumors
A) q-RT-PCR of miR-221 and miR-222 in retrieved tumors treated with unformulated-miR-221 inhibitors or unformulated-NC. The results are shown as averaged miRNA expression after normalization with RNU44 and ΔΔCt calculations as compared to control (miRNA expression in NC treated animals). B) H&E (20-fold magnification) and immunohistochemistry (40-fold magnification) staining of xenografted tumors retrieved from treated animals. C) q-RT-PCR of p27Kip1 and PTEN mRNA levels in treated tumors retrieved from mice. Raw Ct values were normalized to GAPDH housekeeping mRNA and expressed as ΔΔCt values calculated using the comparative cross threshold method. D) Western blot analysis of p27Kip1 and PTEN protein in retrieved tumors from mice treated with miR-221 inhibitors or NC. D-E) Western blot analysis of total AKT and p-AKT (D) and total ERK and p-ERK (E) in retrieved tumors from mice treated with miR-221 inhibitors or NC. Representative experiments are shown in figure. γ-tubulin was the protein loading control.
All figures (7)
Figure 5. Whole Gene Expression Profiles of…
Figure 5. Whole Gene Expression Profiles of MM cells after miR-221/222 knockdown
A) Hierarchical clustering of differentially expressed genes between miR-221/222 inhibitors or NC treated OPM2 cells by Gene 1.0 ST array chip (Affymetrix) and DChip software. Genes that underwent a 0.5-fold change as compared to control, were selected and clustered. Assays performed in triplicate are shown. B) Pathways modulated by miR-221/222 inhibitors in OPM2 MM cells are shown. Analysis was performed by Ingenuity Pathway Analysis on genes significantly modulated (FC>0.5). The bar graphs show pathways most modulated by miR-221/222 inhibitors as compared to control, based on statistical significance (P-value and ratio). The yellow line indicates the threshold of significance. C) Functional network scenario with principal nodes in the most perturbed pathways are showed. Up- and down-regulated genes are indicated in red and green, respectively. Continuous and discontinuous lines represent direct and indirect functional and physical interactions between genes as previously reported.
Figure 6. In vivo anti-tumor activity of…
Figure 6. In vivo anti-tumor activity of miR-221/222 inhibitors in MM xenografted SCID/NOD mice
A) Effects of formulated or unformulated miR-221/222 inhibitors. Subcutaneous OPM2 xenografts were treated every 2 days with 20 μg of formulated(NLE)-miR-221/222 inhibitors or unformulated-miR221/222 inhibitors. As control 2 separate groups of tumor-bearing animals were injected with formulated(NLE)-NC or unformulated-NC. B) Effects of treatments with formulated miR-221 or miR-222 individual inhibitors. Subcutaneous OPM2 xenografts were repeatedly treated every 2 days, with 20 μg of NLE-miR-221 inhibitors, or NLE-miR-222 inhibitors, or NLE-NC. C) Effects of unformulated-miR-221 inhibitors or NC. Subcutaneous OPM2 xenografts were repeatedly treated every 2 days with 20 μg of unformulated-miR-221 inhibitors, or unformulated-NC. The tumor volumes averages of each group +SD are reported. In each experiment P values were calculated for miRNA inhibitors versus scrambled oligonucleotides (NC) and significant P values (P<0.05) are indicated by stars.
Figure 7. miR-221 activity and targets silencing…
Figure 7. miR-221 activity and targets silencing in retrieved MM xenografted tumors
A) q-RT-PCR of miR-221 and miR-222 in retrieved tumors treated with unformulated-miR-221 inhibitors or unformulated-NC. The results are shown as averaged miRNA expression after normalization with RNU44 and ΔΔCt calculations as compared to control (miRNA expression in NC treated animals). B) H&E (20-fold magnification) and immunohistochemistry (40-fold magnification) staining of xenografted tumors retrieved from treated animals. C) q-RT-PCR of p27Kip1 and PTEN mRNA levels in treated tumors retrieved from mice. Raw Ct values were normalized to GAPDH housekeeping mRNA and expressed as ΔΔCt values calculated using the comparative cross threshold method. D) Western blot analysis of p27Kip1 and PTEN protein in retrieved tumors from mice treated with miR-221 inhibitors or NC. D-E) Western blot analysis of total AKT and p-AKT (D) and total ERK and p-ERK (E) in retrieved tumors from mice treated with miR-221 inhibitors or NC. Representative experiments are shown in figure. γ-tubulin was the protein loading control.

References

    1. Anderson KC, Carrasco RD. Pathogenesis of myeloma. Annu Rev Pathol. 2011;6:249–274.
    1. Rajkumar SV. Treatment of multiple myeloma. Nat Rev Clin Oncol. 2011;8(8):479–491.
    1. Morabito F, Recchia AG, Mazzone C, Gentile M. Targeted therapy of multiple myeloma: the changing paradigm at the beginning of the new millennium. Current cancer drug targets. 2012;12(7):743–756.
    1. Lonial S, Mitsiades CS, Richardson PG. Treatment options for relapsed and refractory multiple myeloma. Clin Cancer Res. 2011;17(6):1264–1277.
    1. Kapoor P, Rajkumar SV. Update on risk stratification and treatment of newly diagnosed multiple myeloma. Int J Hematol. 2011
    1. Tassone P, Neri P, Burger R, Di Martino MT, Leone E, Amodio N, Caraglia M, Tagliaferri P. Mouse models as a translational platform for the development of new therapeutic agents in multiple myeloma. Current cancer drug targets. 2012;12(7):814–822.
    1. Rossi M, Di Martino MT, Morelli E, Leotta M, Rizzo A, Grimaldi A, Misso G, Tassone P, Caraglia M. Molecular targets for the treatment of multiple myeloma. Current cancer drug targets. 2012;12(7):757–767.
    1. Neri P, Bahlis NJ. Targeting of adhesion molecules as a therapeutic strategy in multiple myeloma. Current cancer drug targets. 2012;12(7):776–796.
    1. Podar K. Novel targets and derived small molecule inhibitors in multiple myeloma. Current cancer drug targets. 2012;12(7):797–813.
    1. Calimeri T, Battista E, Conforti F, Neri P, Di Martino MT, Rossi M, Foresta U, Piro E, Ferrara F, Amorosi A, Bahlis N, Anderson KC, Munshi N, Tagliaferri P, Causa F, Tassone P. A unique three-dimensional SCID-polymeric scaffold (SCID-synth-hu) model for in vivo expansion of human primary multiple myeloma cells. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2011;25(4):707–711.
    1. Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma. Nature reviews Cancer. 2012;12(5):335–348.
    1. Tassone P, Tagliaferri P, Rossi M, Gaspari M, Terracciano R, Venuta S. Genetics and molecular profiling of multiple myeloma: novel tools for clinical management? Eur J Cancer. 2006;42(11):1530–1538.
    1. Palumbo A, Anderson K. Multiple myeloma. N Engl J Med. 2011;364(11):1046–1060.
    1. Tassone P, Tagliaferri P, Fulciniti MT, Di Martino MT, Venuta S. Novel therapeutic approaches based on the targeting of microenvironment-derived survival pathways in human cancer: experimental models and translational issues. Current pharmaceutical design. 2007;13(5):487–496.
    1. Lionetti M, Biasiolo M, Agnelli L, Todoerti K, Mosca L, Fabris S, Sales G, Deliliers GL, Bicciato S, Lombardi L, Bortoluzzi S, Neri A. Identification of microRNA expression patterns and definition of a microRNA/mRNA regulatory network in distinct molecular groups of multiple myeloma. Blood. 2009;114(25):e20–26.
    1. Benetatos L, Vartholomatos G. Deregulated microRNAs in multiple myeloma. Cancer. 2011
    1. Lionetti M, Agnelli L, Lombardi L, Tassone P, Neri A. MicroRNAs in the pathobiology of multiple myeloma. Curr Cancer Drug Targets. 2012;12(7):823–837.
    1. Tagliaferri P, Rossi M, Di Martino MT, Amodio N, Leone E, Gulla A, Neri A, Tassone P. Promises and Challenges of MicroRNA-based Treatment of Multiple Myeloma. Curr Cancer Drug Targets. 2012;12(7):838–846.
    1. Di Martino MT, Leone E, Amodio N, Foresta U, Lionetti M, Pitari MR, Gallo Cantafio ME, Gulla A, Conforti F, Morelli E, Tomaino V, Rossi M, Negrini M, Ferrarini M, Caraglia M, Shammas MA, et al. Synthetic miR-34a mimics as a novel therapeutic agent for Multiple Myeloma: in vitro and in vivo evidence. Clin Cancer Res. 2012
    1. Amodio N, Di Martino MT, Foresta U, Leone E, Lionetti M, Leotta M, Gulla AM, Pitari MR, Conforti F, Rossi M, Agosti V, Fulciniti M, Misso G, Morabito F, Ferrarini M, Neri A, et al. miR-29b sensitizes multiple myeloma cells to bortezomib-induced apoptosis through the activation of a feedback loop with the transcription factor Sp1. Cell death & disease. 2012;3:e436.
    1. Amodio N, Leotta M, Bellizzi D, Di Martino MT, D'Aquila P, Lionetti M, Fabiani F, Leone E, Gulla AM, Passarino G, Caraglia M, Negrini M, Neri A, Giordano A, Tagliaferri P, Tassone P. DNA-demethylating and anti-tumor activity of synthetic miR-29b mimics in multiple myeloma. Oncotarget. 2012;3(10):1246–1258.
    1. Rossi M, Pitari MR, Amodio N, Di Martino MT, Conforti F, Leone E, Botta C, Paolino FM, Del Giudice T, Iuliano E, Caraglia M, Ferrarini M, Giordano A, Tagliaferri P, Tassone P. miR-29b negatively regulates human osteoclastic cell differentiation and function: Implications for the treatment of multiple myeloma-related bone disease. J Cell Physiol. 2012 Dec;:18.
    1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–297.
    1. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19(1):92–105.
    1. Ambros V. The functions of animal microRNAs. Nature. 2004;431(7006):350–355.
    1. He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5(7):522–531.
    1. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, Croce CM. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A. 2004;101(9):2999–3004.
    1. Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 2009;10(10):704–714.
    1. Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6(4):259–269.
    1. Nohata N, Hanazawa T, Enokida H, Seki N. microRNA-1/133a and microRNA-206/133b clusters: dysregulation and functional roles in human cancers. Oncotarget. 2012;3(1):9–21.
    1. Pekarsky Y, Croce CM. Is miR-29 an oncogene or tumor suppressor in CLL? Oncotarget. 2010;1(3):224–227.
    1. Chang S, Sharan SK. Epigenetic control of an oncogenic microRNA, miR-155, by BRCA1. Oncotarget. 2012;3(1):5–6.
    1. Visone R, Veronese A, Balatti V, Croce CM. MiR-181b: new perspective to evaluate disease progression in chronic lymphocytic leukemia. Oncotarget. 2012;3(2):195–202.
    1. Frenquelli M, Muzio M, Scielzo C, Fazi C, Scarfo L, Rossi C, Ferrari G, Ghia P, Caligaris-Cappio F. MicroRNA and proliferation control in chronic lymphocytic leukemia: functional relationship between miR-221/222 cluster and p27. Blood. 2010;115(19):3949–3959.
    1. le Sage C, Nagel R, Egan DA, Schrier M, Mesman E, Mangiola A, Anile C, Maira G, Mercatelli N, Ciafre SA, Farace MG, Agami R. Regulation of the p27(Kip1) tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. The EMBO journal. 2007;26(15):3699–3708.
    1. Medina R, Zaidi SK, Liu CG, Stein JL, van Wijnen AJ, Croce CM, Stein GS. MicroRNAs 221 and 222 bypass quiescence and compromise cell survival. Cancer Res. 2008;68(8):2773–2780.
    1. Garofalo M, Di Leva G, Romano G, Nuovo G, Suh SS, Ngankeu A, Taccioli C, Pichiorri F, Alder H, Secchiero P, Gasparini P, Gonelli A, Costinean S, Acunzo M, Condorelli G, Croce CM. miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell. 2009;16(6):498–509.
    1. Chun-Zhi Z, Lei H, An-Ling Z, Yan-Chao F, Xiao Y, Guang-Xiu W, Zhi-Fan J, Pei-Yu P, Qing-Yu Z, Chun-Sheng K. MicroRNA-221 and microRNA-222 regulate gastric carcinoma cell proliferation and radioresistance by targeting PTEN. BMC cancer. 2010;10
    1. Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, Terris B, Mazzaferro V, Lowe SW, Croce CM, Dejean A. miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci U S A. 2010;107(1):264–269.
    1. Galardi S, Mercatelli N, Giorda E, Massalini S, Frajese GV, Ciafre SA, Farace MG. miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J Biol Chem. 2007;282(32):23716–23724.
    1. Zhang C, Kang C, You Y, Pu P, Yang W, Zhao P, Wang G, Zhang A, Jia Z, Han L, Jiang H. Co-suppression of miR-221/222 cluster suppresses human glioma cell growth by targeting p27kip1 in vitro and in vivo. International journal of oncology. 2009;34(6):1653–1660.
    1. Wiggins JF, Ruffino L, Kelnar K, Omotola M, Patrawala L, Brown D, Bader AG. Development of a lung cancer therapeutic based on the tumor suppressor microRNA-34. Cancer Res. 2010;70(14):5923–5930.
    1. Trang P, Wiggins JF, Daige CL, Cho C, Omotola M, Brown D, Weidhaas JB, Bader AG, Slack FJ. Systemic delivery of tumor suppressor microRNA mimics using a neutral lipid emulsion inhibits lung tumors in mice. Mol Ther. 2011;19(6):1116–1122.
    1. Visone R, Russo L, Pallante P, De Martino I, Ferraro A, Leone V, Borbone E, Petrocca F, Alder H, Croce CM, Fusco A. MicroRNAs (miR)-221 and miR-222, both overexpressed in human thyroid papillary carcinomas, regulate p27Kip1 protein levels and cell cycle. Endocrine-related cancer. 2007;14(3):791–798.
    1. Miller TE, Ghoshal K, Ramaswamy B, Roy S, Datta J, Shapiro CL, Jacob S, Majumder S. MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J Biol Chem. 2008;283(44):29897–29903.
    1. Fu X, Wang Q, Chen J, Huang X, Chen X, Cao L, Tan H, Li W, Zhang L, Bi J, Su Q, Chen L. Clinical significance of miR-221 and its inverse correlation with p27Kip(1) in hepatocellular carcinoma. Molecular biology reports. 2011;38(5):3029–3035.
    1. Garofalo M, Quintavalle C, Di Leva G, Zanca C, Romano G, Taccioli C, Liu CG, Croce CM, Condorelli G. MicroRNA signatures of TRAIL resistance in human non-small cell lung cancer. Oncogene. 2008;27(27):3845–3855.
    1. Fornari F, Gramantieri L, Ferracin M, Veronese A, Sabbioni S, Calin GA, Grazi GL, Giovannini C, Croce CM, Bolondi L, Negrini M. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene. 2008;27(43):5651–5661.
    1. Quintavalle C, Garofalo M, Zanca C, Romano G, Iaboni M, del Basso De Caro M, Martinez-Montero JC, Incoronato M, Nuovo G, Croce CM, Condorelli G. miR-221/222 overexpession in human glioblastoma increases invasiveness by targeting the protein phosphate PTPmu. Oncogene. 2012;31(7):858–868.
    1. Cesario RM, Stone J, Yen WC, Bissonnette RP, Lamph WW. Differentiation and growth inhibition mediated via the RXR:PPARgamma heterodimer in colon cancer. Cancer Lett. 2006;240(2):225–233.
    1. Albino D, Longoni N, Curti L, Mello-Grand M, Pinton S, Civenni G, Thalmann G, D'Ambrosio G, Sarti M, Sessa F, Chiorino G, Catapano CV, Carbone GM. ESE3/EHF controls epithelial cell differentiation and its loss leads to prostate tumors with mesenchymal and stem-like features. Cancer Res. 2012;72(11):2889–2900.
    1. Liu X, Yu J, Jiang L, Wang A, Shi F, Ye H, Zhou X. MicroRNA-222 regulates cell invasion by targeting matrix metalloproteinase 1 (MMP1) and manganese superoxide dismutase 2 (SOD2) in tongue squamous cell carcinoma cell lines. Cancer genomics & proteomics. 2009;6(3):131–139.
    1. Catley L, Tai YT, Shringarpure R, Burger R, Son MT, Podar K, Tassone P, Chauhan D, Hideshima T, Denis L, Richardson P, Munshi NC, Anderson KC. Proteasomal degradation of topoisomerase I is preceded by c-Jun NH2-terminal kinase activation, Fas up-regulation, and poly(ADP-ribose) polymerase cleavage in SN38-mediated cytotoxicity against multiple myeloma. Cancer research. 2004;64(23):8746–8753.
    1. Hideshima T, Bergsagel PL, Kuehl WM, Anderson KC. Advances in biology of multiple myeloma: clinical applications. Blood. 2004;104(3):607–618.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–408.
    1. Guzzi PH, Di Martino MT, Tradigo G, Veltri P, Tassone P, Tagliaferri P, Cannataro M. Automatic summarisation and annotation of microarray data. Soft Comput. 2011;15(8):1505–1512.
    1. Li C, Wong WH. Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc Natl Acad Sci U S A. 2001;98(1):31–36.
    1. Tassone P, Di Martino MT, Ventura M, Pietragalla A, Cucinotto I, Calimeri T, Bulotta A, Neri P, Caraglia M, Tagliaferri P. Loss of BRCA1 function increases the antitumor activity of cisplatin against human breast cancer xenografts in vivo. Cancer biology & therapy. 2009;8(7):648–653.
    1. Neri P, Tagliaferri P, Di Martino MT, Calimeri T, Amodio N, Bulotta A, Ventura M, Eramo PO, Viscomi C, Arbitrio M, Rossi M, Caraglia M, Munshi NC, Anderson KC, Tassone P. In vivo anti-myeloma activity and modulation of gene expression profile induced by valproic acid, a histone deacetylase inhibitor. British journal of haematology. 2008;143(4):520–531.

Source: PubMed

3
Tilaa