MiR-126 and miR-126* regulate shear-resistant firm leukocyte adhesion to human brain endothelium

Camilla Cerutti, Laura J Edwards, Helga E de Vries, Basil Sharrack, David K Male, Ignacio A Romero, Camilla Cerutti, Laura J Edwards, Helga E de Vries, Basil Sharrack, David K Male, Ignacio A Romero

Abstract

Leukocyte adhesion to brain endothelial cells, the blood-brain barrier main component, is a critical step in the pathogenesis of neuroinflammatory diseases such as multiple sclerosis (MS). Leukocyte adhesion is mediated mainly by selectins, cell adhesion molecules and chemokines induced by pro-inflammatory cytokines such as TNFα and IFNγ, but the regulation of this process is not fully clear. This study investigated the regulation of firm leukocyte adhesion to human brain endothelium by two different brain endothelial microRNAs (miRs), miR-126 and miR-126*, that are downregulated by TNFα and IFNγ in a human brain endothelial cell line, hCMEC/D3. Using a leukocyte adhesion in vitro assay under shear forces mimicking blood flow, we observed that reduction of endothelial miR-126 and miR-126* enhanced firm monocyte and T cell adhesion to hCMEC/D3 cells, whereas their increased expression partially prevented THP1, Jurkat and primary MS patient-derived PBMC firm adhesion. Furthermore, we observed that miR-126* and miR-126 downregulation increased E-selectin and VCAM1, respectively, while miR-126 overexpression reduced VCAM1 and CCL2 expression by hCMEC/D3 cells, suggesting that these miRs regulate leukocyte adhesion by modulating the expression of adhesion-associated endothelial mRNA targets. Hence, human brain endothelial miR-126 and miR-126* could be used as a therapeutic tool to reduce leukocyte adhesion and thus reduce neuroinflammation.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. TNFα + IFNγ increase E-selectin,…
Figure 1. TNFα + IFNγ increase E-selectin, ICAM1 and VCAM1 expression, enhance firm leukocyte adhesion and downregulate miR-126 and miR-126* expression in hCMEC/D3 cells.
hCMEC/D3 cell monolayers were treated with a combination of cytokines (TNFα + IFNγ) (A) at different concentrations (0, 0.1, 1, 10 ng/ml) or (BD,F) at 1 ng/ml or left without (w/o) for 24 h. (A) VCAM1, ICAM1 and E-selectin expression levels were quantified by ELISA. (B) VCAM1 expression was quantified by immunofluorescence and expressed as Integrated Density in arbitrary units (A.U.). (C) Numbers of shear-resistant firmly adhered Jurkat and THP1 cells to hCMEC/D3 monolayer per field of view (FOV) (640 × 480 μm). (D) Representative images of shear-resistant firmly adhered fluorescently labelled Jurkat T cells (small white rounded cells) to unstimulated (left) or cytokine treated (right) hCMEC/D3 cells (monolayer of spindle shaped cells in bright field) per FOV (640 × 480 μm). (E) Heatmap representing miR-126 and miR-126* expression in unstimulated hCMEC/D3 cells and after stimulation (10 ng/ml of TNFα + IFNγ) for 24 h (n = 3). Individual repeats are shown in the heatmap. Blue indicates under expression, red overexpression, and intensity of color indicates relative change. Rows were colored using a z-score derived from a gene’s expression across all samples (row z-score). Data from Geo accession GSE21350 (Platform GPL14767) of miR-126 and miR-126* are represented. (F) Relative miR-126 and miR-126* level expression measured by qRT2-PCR from total RNA. The small nuclear RNA U6 was used as internal control. Experiments were carried out three times to four times with (A,B,F) three replicates (C) five FOV. Data are mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, *compared to untreated (w/o TNFα + IFNγ).
Figure 2. Human brain endothelial miR-126 downregulation…
Figure 2. Human brain endothelial miR-126 downregulation enhances shear-resistant firmly adhered Jurkat and THP1 cells to hCMEC/D3 cells.
hCMEC/D3 cell were transfected with control Scrambled Anti-miR (grey) or with Anti-miR-126 (black) followed by treatment with cytokines (TNFα + IFNγ, 1 ng/ml) for 24 h or left unstimulated. (A) Relative miR-126 level expression measured by qRT2-PCR from total RNA. The small nuclear RNA U6 was used as internal control. (B,C) Numbers of shear-resistant firmly adhered Jurkat T cells and THP1 monocytes to (B) unstimulated and (C) cytokine-stimulated hCMEC/D3 cells per field of view (FOV) (640 × 480 μm). Experiments were carried out three to five times (A) three replicates (B,C) five FOVs. Data are mean ± SEM. #P < 0.05, **P < 0.01, #compared to unstimulated.
Figure 3. miR-126 upregulation decreases monocytic, T…
Figure 3. miR-126 upregulation decreases monocytic, T cell, PBMC and MS-derived PBMC firm adhesion to hCMEC/D3 cells.
hCMEC/D3 cells were transfected with control Scrambled Pre-miR (grey) or with Pre-miR-126 (black) followed by treatment with cytokines (TNFα + IFNγ, 1 ng/ml) for 24 h or left unstimulated. (A) Relative miR-126 level expression measured by qRT2-PCR from total RNA. The small nuclear RNA U6 was used as internal control. (B,C) Numbers of shear-resistant firmly adhered Jurkat T cells and THP1 monocytes to (B) unstimulated and (C) cytokine-stimulated hCMEC/D3 monolayers per field of view (FOV) (640 × 480 μm). (D) Information of the three PBMC healthy donors and multiple sclerosis (MS) patient PBMC donors; pathological features, relapsing-remitting MS (RRMS), secondary progressive MS (SPMS), primary progressive (PPMS). (E,F) Numbers of shear-resistant firmly adhered (E) healthy donor PBMC and (F) MS patient derived PBMC to cytokine-stimulated hCMEC/D3 monolayers per field of view (FOV) (640 × 480 μm). (G) Representative images of shear-resistant firmly adhered healthy donor PBMC and MS patient derived PBMC MS to control scrambled Pre-miR or Pre-miR-126 transfected and cytokine-treated hCMEC/D3 cells (monolayer of spindle shaped cells in bright field) per FOV (640 × 480 μm). Experiments were carried out (B,C) three times to five times with five FOVs and (E,F) with ten FOVs. Data are mean ± SEM. *,#P < 0.05, **P < 0.01, ***P < 0.001, #compared to unstimulated.
Figure 4. miR-126* downregulation regulates THP1 monocyte…
Figure 4. miR-126* downregulation regulates THP1 monocyte and Jurkat T cell firm adhesion to hCMEC/D3 cells.
hCMEC/D3 cell were transfected with control Scrambled Anti-miR (grey) or with Anti-miR-126* (black) followed by treatment with cytokines (TNFα + IFNγ, 1 ng/ml) for 24 h or left unstimulated. (A) Relative miR-126* level expression measured by qRT2-PCR from total RNA. The small nuclear RNA U6 was used as internal control. (B,C) Numbers of shear-resistant firmly adhered (B) THP1 monocytes and (C) Jurkat T cells to unstimulated and cytokine-stimulated hCMEC/D3 cells per field of view (FOV) (640 × 480 μm). Experiments were carried out three times with (A) two replicates (B,C) five FOVs. Data are mean ± SEM. #P < 0.05, **P < 0.01, ***,###P < 0.001, #compared to unstimulated.
Figure 5. miR-126* upregulation decreases shear-resistant firmly…
Figure 5. miR-126* upregulation decreases shear-resistant firmly adhered Jurkat and THP1 cells to hCMEC/D3 cells.
hCMEC/D3 cells were transfected with control Scrambled Pre-miR (grey) or with Pre-miR-126 (black) (A) Relative miR-126* level expression measured by qRT2-PCR from total RNA. The small nuclear RNA U6 was used as internal control. (B,C) Numbers of shear-resistant firmly adhered THP1 monocytes and Jurkat T cells to (B) unstimulated and (C) cytokine-stimulated hCMEC/D3 cells per field of view (FOV) (640 × 480 μm). Experiments were carried out three times with five FOVs. Data are mean ± SEM. #P < 0.05, **P < 0.01, ***,###P < 0.001, #compared to unstimulated.
Figure 6. Identification of miR-126 and miR-126*…
Figure 6. Identification of miR-126 and miR-126* adhesion-related putative gene targets in silico.
(A) Stem loop of the primary precursor (Pri-miR) structure of human miR-126 and miR-126* and the mature hsa-miR-126 and has-miR-126* sequences. (B) List of adhesion-associated hsa-miR-126 and hsa-miR-126* predicted targets in silico. (C) Sequence alignment of the predicted duplex formation between mature miRs and their putative gene target mRNA selected for further study in hCMEC/D3 cells. (D) Graphic representation of the experimental design used to study the predicted target at protein level, the same experimental well was used to study membrane associated protein and secreted protein.
Figure 7. Brain endothelial miR-126 modulates VCAM1…
Figure 7. Brain endothelial miR-126 modulates VCAM1 and CCL2 in hCMEC/D3 cells.
(AD) hCMEC/D3 cells were transfected with control Scrambled Pre-miR or Pre-miR-126 or control Scrambled Anti-miR or Anti-miR-126 (C,D) followed by treatment with cytokines (TNFα + IFNγ, 1 ng/ml) for 24 h or (A,B) left unstimulated. (A,C) Membrane-associated VCAM1 expression was quantified by ELISA and (B,D) secreted CCL2 (MCP1) was measured by capture ELISA. Experiments were carried out (A,C) four or (B,D) three times with three replicates each. Data are mean ± SEM. *P < 0.05, ###P < 0.001, #compared to unstimulated.
Figure 8. Brain endothelial miR-126* modulates E-selectin…
Figure 8. Brain endothelial miR-126* modulates E-selectin in hCMEC/D3 cells.
(A–D) hCMEC/D3 cells were transfected with control Scrambled Pre-miR or Pre-miR-126 or control Scrambled Anti-miR or Anti-miR-126 (C,D) followed by treatment with cytokines (TNFα + IFNγ, 1 ng/ml) for 24 h or (A,B) left unstimulated. (A,C) Membrane-associated E-selectin (CD62E) expression was quantified by ELISA and (B,D) secreted CCL7 (MCP3) was measured by capture ELISA. Experiments were carried out three times with three replicates each. Data are mean ± SEM. *P < 0.05, **P < 0.01, ###P < 0.001, #compared to unstimulated.

References

    1. Engelhardt B. & Ransohoff R. M. Capture, crawl, cross: the T cell code to breach the blood-brain barriers. Trends Immunol 33, 579–589, doi: 10.1016/j.it.2012.07.004 (2012).
    1. Ousman S. S. & Kubes P. Immune surveillance in the central nervous system. Nat Neurosci 15, 1096–1101, doi: 10.1038/nn.3161 (2012).
    1. McFarland H. F. & Martin R. Multiple sclerosis: a complicated picture of autoimmunity. Nat Immunol 8, 913–919, doi: 10.1038/ni1507 (2007).
    1. Owens T., Bechmann I. & Engelhardt B. Perivascular spaces and the two steps to neuroinflammation. J Neuropathol Exp Neurol 67, 1113–1121, doi: 10.1097/NEN.0b013e31818f9ca8 (2008).
    1. Hughes C. C., Male D. K. & Lantos P. L. Adhesion of lymphocytes to cerebral microvascular cells: effects of interferon-gamma, tumour necrosis factor and interleukin-1. Immunology 64, 677–681 (1988).
    1. Sharief M. K. & Hentges R. Association between tumor necrosis factor-alpha and disease progression in patients with multiple sclerosis. N Engl J Med 325, 467–472, doi: 10.1056/nejm199108153250704 (1991).
    1. Sospedra M. & Martin R. Immunology of multiple sclerosis. Annual review of immunology 23, 683–747, doi: 10.1146/annurev.immunol.23.021704.115707 (2005).
    1. Engelhardt B. Regulation of immune cell entry into the central nervous system. Results Probl Cell Differ 43, 259–280 (2006).
    1. Lucchinetti C. F., Parisi J. & Bruck W. The pathology of multiple sclerosis. Neurologic clinics 23, 77–105, vi, doi: 10.1016/j.ncl.2004.09.002 (2005).
    1. McManus C. et al.. MCP-1, MCP-2 and MCP-3 expression in multiple sclerosis lesions: an immunohistochemical and in situ hybridization study. J Neuroimmunol 86, 20–29, doi: 10.1016/S0165-5728(98)00002-2 (1998).
    1. Olson T. S. & Ley K. Chemokines and chemokine receptors in leukocyte trafficking. Am J Physiol Regul Integr Comp Physiol 283, R7–28, doi: 10.1152/ajpregu.00738.2001 (2002).
    1. Cerutti C. et al.. MicroRNA-155 contributes to shear-resistant leukocyte adhesion to human brain endothelium in vitro. Fluids Barriers CNS 13, 8, doi: 10.1186/s12987-016-0032-3 (2016).
    1. Bartel D. P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233, doi: 10.1016/j.cell.2009.01.002 (2009).
    1. Thamilarasan M., Koczan D., Hecker M., Paap B. & Zettl U. K. MicroRNAs in multiple sclerosis and experimental autoimmune encephalomyelitis. Autoimmun Rev 11, 174–179, doi: 10.1016/j.autrev.2011.05.009 (2012).
    1. Rom S., Dykstra H., Zuluaga-Ramirez V., Reichenbach N. L. & Persidsky Y. miR-98 and let-7g* protect the blood-brain barrier under neuroinflammatory conditions. J Cereb Blood Flow Metab 35, 1957–1965, doi: 10.1038/jcbfm.2015.154 (2015).
    1. Wu D. et al.. Brain endothelial miR-146a negatively modulates T-cell adhesion through repressing multiple targets to inhibit NF-kappaB activation. J Cereb Blood Flow Metab 35, 412–423, doi: 10.1038/jcbfm.2014.207 (2015).
    1. Fish J. E. et al.. miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell 15, 272–284, doi: 10.1016/j.devcel.2008.07.008 (2008).
    1. Harris T. A., Yamakuchi M., Ferlito M., Mendell J. T. & Lowenstein C. J. MicroRNA-126 regulates endothelial expression of vascular cell adhesion molecule 1. Proc Natl Acad Sci USA 105, 1516–1521, doi: 10.1073/pnas.0707493105 (2008).
    1. McCall M. N. et al.. MicroRNA profiling of diverse endothelial cell types. BMC Med Genomics 4, 78, doi: 10.1186/1755-8794-4-78 (2011).
    1. Wang S. et al.. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell 15, 261–271, doi: 10.1016/j.devcel.2008.07.002 (2008).
    1. Taverna S. et al.. Exosomal shuttling of miR-126 in endothelial cells modulates adhesive and migratory abilities of chronic myelogenous leukemia cells. Mol Cancer 13, 169, doi: 10.1186/1476-4598-13-169 (2014).
    1. Yang J. S. et al.. Widespread regulatory activity of vertebrate microRNA* species. Rna 17, 312–326, doi: 10.1261/rna.2537911 (2011).
    1. Huang X. et al.. Regulated expression of microRNAs-126/126* inhibits erythropoiesis from human embryonic stem cells. Blood 117, 2157–2165, doi: 10.1182/blood-302711 (2010) (2011).
    1. Schober A. et al.. MicroRNA-126-5p promotes endothelial proliferation and limits atherosclerosis by suppressing Dlk1. Nat Med 20, 368–376, doi: 10.1038/nm.3487 (2014).
    1. Meister J. & Schmidt M. H. miR-126 and miR-126*: new players in cancer. ScientificWorldJournal 10, 2090–2100, doi: 10.1100/tsw.2010.198 (2010).
    1. Musiyenko A., Bitko V. & Barik S. Ectopic expression of miR-126*, an intronic product of the vascular endothelial EGF-like 7 gene, regulates prostein translation and invasiveness of prostate cancer LNCaP cells. J Mol Med (Berl) 86, 313–322, doi: 10.1007/s00109-007-0296-9 (2008).
    1. Felli N. et al.. miR-126&126* Restored Expressions Play a Tumor Suppressor Role by Directly Regulating ADAM9 and MMP7 in Melanoma. PLoS One 8, e56824; doi: 10.1371/pone.0056824 (2013).
    1. Zhang Y. et al.. miR-126 and miR-126(*) repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol 15, 284–294, doi: 10.1038/ncb2690 (2013).
    1. Poissonnier L., Villain G., Soncin F. & Mattot V. miR126-5p repression of ALCAM and SetD5 in endothelial cells regulates leucocyte adhesion and transmigration. Cardiovasc Res 102, 436–447, doi: 10.1093/cvr/cvu040 (2014).
    1. Weksler B. B. et al.. Blood-brain barrier-specific properties of a human adult brain endothelial cell line. FASEB J. 04-3458fje, doi: 10.1096/fj.04-3458fje (2005).
    1. Wong D. & Dorovini-Zis K. Regualtion by cytokines and lipopolysaccharide of E-selectin expression by human brain microvessel endothelial cells in primary culture. J Neuropathol Exp Neurol 55, 225–235 (1996).
    1. de Fougerolles A. R., Stacker S. A., Schwarting R. & Springer T. A. Characterization of ICAM-2 and evidence for a third counter-receptor for LFA-1. J Exp Med 174, 253–267 (1991).
    1. Nortamo P. et al.. The expression of human intercellular adhesion molecule-2 is refractory to inflammatory cytokines. Eur J Immunol 21, 2629–2632, doi: 10.1002/eji.1830211049 (1991).
    1. McLaughlin F. et al.. Tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta down-regulate intercellular adhesion molecule (ICAM)-2 expression on the endothelium. Cell Adhes Commun 6, 381–400 (1998).
    1. Lopez-Ramirez M. A. et al.. MicroRNA-155 negatively affects blood-brain barrier function during neuroinflammation. Faseb j 28, 2551–2565, doi: 10.1096/fj.13-248880 (2014).
    1. Battistini L. et al.. CD8+ T cells from patients with acute multiple sclerosis display selective increase of adhesiveness in brain venules: a critical role for P-selectin glycoprotein ligand-1. Blood 101, 4775–4782, doi: 10.1182/blood-2002-10-3309 (2003).
    1. Rudolph H. et al.. Postarrest stalling rather than crawling favors CD8+ over CD4+ T-cell migration across the blood-brain barrier under flow in vitro. European journal of immunology, doi: 10.1002/eji.201546251 (2016).
    1. Sun C. et al.. IRF-1 and miRNA126 modulate VCAM-1 expression in response to a high-fat meal. Circ Res 111, 1054–1064, doi: 10.1161/circresaha.112.270314 (2012).
    1. Asgeirsdottir S. A. et al.. MicroRNA-126 contributes to renal microvascular heterogeneity of VCAM-1 protein expression in acute inflammation. Am J Physiol Renal Physiol 302, F1630–1639, doi: 10.1152/ajprenal.00400.2011 (2012).
    1. Salvucci O. et al.. MicroRNA126 contributes to granulocyte colony-stimulating factor-induced hematopoietic progenitor cell mobilization by reducing the expression of vascular cell adhesion molecule 1. Haematologica 97, 818–826, doi: 10.3324/haematol.2011.056945 (2012).
    1. Arner E. et al.. Adipose tissue microRNAs as regulators of CCL2 production in human obesity. Diabetes 61, 1986–1993, doi: 10.2337/db11-1508 (2012).
    1. Zhang Y. et al.. miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol 15, 284–294, doi: 10.1038/ncb2690 (2013).
    1. Chen J. et al.. MiR-126 Contributes to Human Umbilical Cord Blood Cell-Induced Neurorestorative Effects After Stroke in Type-2 Diabetic Mice. Stem Cells 34, 102–113, doi: 10.1002/stem.2193 (2016).
    1. Zernecke A. et al.. Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal 2, ra81, doi: 10.1126/scisignal.2000610 (2009).
    1. Minagar A. & Alexander J. S. Blood-brain barrier disruption in multiple sclerosis. Mult Scler 9, 540–549 (2003).
    1. Takeshita Y. & Ransohoff R. M. Inflammatory cell trafficking across the blood-brain barrier: chemokine regulation and in vitro models. Immunol Rev 248, 228–239, doi: 10.1111/j.1600-065X.2012.01127.x (2012).
    1. Lopez-Ramirez M. A. et al.. Role of caspases in cytokine-induced barrier breakdown in human brain endothelial cells. J Immunol 189, 3130–3139, doi: 10.4049/jimmunol.1103460 (2012).
    1. Soilu-Hanninen M. et al.. Progressive multifocal leukoencephalopathy as a complication of natalizumab therapy. Duodecim 129, 765–770 (2013).
    1. Lim L. P. et al.. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433, 769–773, doi: 10.1038/nature03315 (2005).
    1. Guo H., Ingolia N. T., Weissman J. S. & Bartel D. P. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 466, 835–840, doi: 10.1038/nature09267 (2010).
    1. Baek D. et al.. The impact of microRNAs on protein output. Nature 455, 64–71, doi: 10.1038/nature07242 (2008).
    1. Inui M., Martello G. & Piccolo S. MicroRNA control of signal transduction. Nat Rev Mol Cell Biol 11, 252–263, doi: 10.1038/nrm2868 (2010).
    1. Oglesby I. K. et al.. miR-126 is downregulated in cystic fibrosis airway epithelial cells and regulates TOM1 expression. J Immunol 184, 1702–1709, doi: 10.4049/jimmunol.0902669 (2010).
    1. Bai Y. et al.. MicroRNA-126 inhibits ischemia-induced retinal neovascularization via regulating angiogenic growth factors. Exp Mol Pathol 91, 471–477, doi: 10.1016/j.yexmp.2011.04.016 (2011).
    1. Yanaihara N. et al.. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 9, 189–198, doi: 10.1016/j.ccr.2006.01.025 (2006).
    1. Harris T. A., Yamakuchi M., Kondo M., Oettgen P. & Lowenstein C. J. Ets-1 and Ets-2 regulate the expression of microRNA-126 in endothelial cells. Arterioscler Thromb Vasc Biol 30, 1990–1997, doi: 10.1161/atvbaha.110.211706 (2010).
    1. Goetze S. et al.. TNFalpha induces expression of transcription factors c-fos, Egr-1, and Ets-1 in vascular lesions through extracellular signal-regulated kinases 1/2. Atherosclerosis 159, 93–101 (2001).
    1. Russell L. & Garrett-Sinha L. A. Transcription factor Ets-1 in cytokine and chemokine gene regulation. Cytokine 51, 217–226, doi: 10.1016/j.cyto.2010.03.006 (2010).
    1. Hu S., Zhu W., Zhang L. F., Pei M. & Liu M. F. MicroRNA-155 broadly orchestrates inflammation-induced changes of microRNA expression in breast cancer. Cell research 24, 254–257, doi: 10.1038/cr.2013.137 (2014).
    1. Westmuckett A. D., Thacker K. M. & Moore K. L. Tyrosine sulfation of native mouse Psgl-1 is required for optimal leukocyte rolling on P-selectin in vivo. PLoS One 6, e20406, doi: 10.1371/journal.pone.0020406 (2011).
    1. Nishimura Y. et al.. Human P-selectin glycoprotein ligand-1 is a functional receptor for enterovirus 71. Nat Med 15, 794–797, doi: 10.1038/nm.1961 (2009).
    1. Ko Y. C. et al.. Endothelial CD200 is heterogeneously distributed, regulated and involved in immune cell-endothelium interactions. J Anat 214, 183–195, doi: 10.1111/j.1469-7580.2008.00986.x (2009).
    1. Cayrol R. et al.. Activated leukocyte cell adhesion molecule promotes leukocyte trafficking into the central nervous system. Nat Immunol 9, 137–145, doi: 10.1038/ni1551 (2008).
    1. Broderick J. A. & Zamore P. D. MicroRNA therapeutics. Gene Ther 18, 1104–1110; doi: 10.1038/gt.2011.50 (2011).
    1. Collison A. et al.. Altered expression of microRNA in the airway wall in chronic asthma: miR-126 as a potential therapeutic target. BMC Pulm Med 11, doi: 10.1186/1471-2466-11-29 (2011).
    1. Kota J. et al.. Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell 137, 1005–1017; doi: 10.1016/j.cell.2009.04.021 (2009).
    1. Endo-Takahashi Y. et al.. Systemic delivery of miR-126 by miRNA-loaded Bubble liposomes for the treatment of hindlimb ischemia. Sci Rep 4, 3883, doi: 10.1038/srep03883 (2014).
    1. Ma S. et al.. E-selectin-targeting delivery of microRNAs by microparticles ameliorates endothelial inflammation and atherosclerosis. Sci Rep 6, 22910, doi: 10.1038/srep22910 (2016).
    1. Male D. K., Pryce G. & Hughes C. C. Antigen presentation in brain: MHC induction on brain endothelium and astrocytes compared. Immunology 60, 453–459 (1987).
    1. Livak K. J. & Schmittgen T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402–408, doi: 10.1006/meth.2001.1262 (2001).

Source: PubMed

3
订阅