miR-142-3p Regulates BDNF Expression in Activated Rodent Microglia Through Its Target CAMK2A

Neelima Gupta, Shweta Jadhav, Kai-Leng Tan, Genevieve Saw, Karthik Babu Mallilankaraman, S Thameem Dheen, Neelima Gupta, Shweta Jadhav, Kai-Leng Tan, Genevieve Saw, Karthik Babu Mallilankaraman, S Thameem Dheen

Abstract

Microglia, the innate immune effector cells of the mammalian central nervous system (CNS), are involved in the development, homeostasis, and pathology of CNS. Microglia become activated in response to various insults and injuries and protect the CNS by phagocytosing the invading pathogens, dead neurons, and other cellular debris. Recent studies have demonstrated that the epigenetic mechanisms ensure the coordinated regulation of genes involved in microglial activation. In this study, we performed a microRNA (miRNA) microarray in activated primary microglia derived from rat pup's brain and identified differentially expressed miRNAs targeting key genes involved in cell survival, apoptosis, and inflammatory responses. Interestingly, miR-142-3p, one of the highly up-regulated miRNAs in microglia upon lipopolysaccharide (LPS)-mediated activation, compared to untreated primary microglia cells was predicted to target Ca2+/calmodulin dependent kinase 2a (CAMK2A). Further, luciferase reporter assay confirmed that miR-142-3p targets the 3'UTR of Camk2a. CAMK2A has been implicated in regulating the expression of brain-derived neurotrophic factor (BDNF) and long-term potentiation (LTP), a cellular mechanism underlying memory and learning. Given this, this study further focused on understanding the miR-142-3p mediated regulation of the CAMK2A-BDNF pathway via Cyclic AMP-responsive element-binding protein (CREB) in activated microglia. The results revealed that CAMK2A was downregulated in activated microglia, suggesting an inverse relationship between miR-142-3p and Camk2a in activated microglia. Overexpression of miR-142-3p in microglia was found to decrease the expression of CAMK2A and subsequently BDNF through regulation of CREB phosphorylation. Functional analysis through shRNA-mediated stable knockdown of CAMK2A in microglia confirmed that the regulation of BDNF by miR-142-3p is via CAMK2A. Overall, this study provides a database of differentially expressed miRNAs in activated primary microglia and reveals that microglial miR-142-3p regulates the CAMK2A-CREB-BDNF pathway which is involved in synaptic plasticity.

Keywords: Ca2+/calmodulin dependent kinase 2a (CAMK2A); brain-derived neurotrophic factor (BDNF); microRNA; microRNA-142 (miR-142-3p/5p); microarray; microglia BV2; rodent primary microglia.

Copyright © 2020 Gupta, Jadhav, Tan, Saw, Mallilankaraman and Dheen.

Figures

Figure 1
Figure 1
Global miRNA expression profiling shows the differential expression of miRNAs in activated microglia. (A) Heat map generated from microRNA microarray shows the result of differential miRNAs expression in control vs. lipopolysaccharide (LPS)-treated primary microglia. Each column represents a biological sample while each row represents a miRNA. The color scale represents the relative expression level of each miRNA in each sample as compared to the reference channel. Red color indicates an expression that is higher than the reference channel, while green color indicates an expression that is below the reference. (B) Volcano plot illustrating the relation between the logarithm of the p-values and the log fold change of miRNAs differentially expressed in control and LPS-activated microglia. The top selected miRNAs are marked with annotation on the plot and tabulated on the right. (C) Quantitative RT-PCR analysis of few differentially expressed miRNAs found in miRNA microarray in Control vs. LPS activated primary microglia. Results are represented as fold change for untreated controls. Statistical analysis was carried out using ANOVA with post hoc Tukey test. Data represented as mean ± SD, (n = 5), ***p < 0.001; **p < 0.01; *p < 0.05.
Figure 2
Figure 2
Ingenuity pathway analysis (IPA) analysis of differentially expressed miRNAs in activated primary microglia compared to control microglial cells. (A,B) IPA showing up- (red) and down-regulation (green) of miRNAs and their target gene networks such as NF-κB (A) and Akt (B) in LPS-activated primary microglial cells when compared to control.
Figure 3
Figure 3
miR-142-3p/5p upregulates in activated microglia and targets CAMK2A. (A) Histogram depicts a significant upregulation of miR-142-3p and miR-142-5p in activated microglia upon LPS treatment for 6 h. Data represented as mean ± SD, (n = 5), Students t-test, *p < 0.05. (B) Bioinformatics analysis predicted that miR-142-3p and miR-142-5p putatively target 3′UTR of Camk2a. The seed region and the target site type have been highlighted (TargetScan and Miranda software was used to predict miRNA targets. Image acquired from http://www.microrna.org/). (C) Histogram depicting a significant decrease in mRNA expression levels of Camk2a in activated microglia upon LPS treatment for 6 h, suggesting an inverse relationship between miR-142-3p/5p and its target gene, Camk2a. Data represented as mean ± SD, (n = 4), Students t-test, ***p < 0.001.
Figure 4
Figure 4
Downregulation of CAMK2A-BDNF pathway proteins in LPS-activated primary microglia. (A) Immunoblots of CAMK2A (50 kDa), phosphorylation of CREB (pCREB; 43 kDa), BDNF (15 kDa) in LPS activated primary microglia show significant downregulation. Total cyclic AMP-responsive element-binding protein (CREB) and β-actin show no difference between control and activated microglia. (B) Histogram shows the significant downregulation of CAMK2A, pCREB, and BDNF in microglia treated with LPS for 6 h. Immunoblot data has been quantified and normalized to β-actin. Phospho-CREB was then normalized to total CREB. Data represented as mean ± SD, (n = 3), Students t-test, *p < 0.05. (C) Immunofluorescence analysis shows that CAMK2A (red) expression appears to be decreased in primary microglia treated with LPS for 6 h when compared to controls. (D) Immunofluorescence analysis shows that BDNF (red) expression also appears to be decreased in primary microglia treated with LPS for 6 h when compared to controls. Lectin (green) used as microglial markers. Nuclei are stained with DAPI (blue), (n = 3), Scale bars = 20 μm.
Figure 5
Figure 5
miR-142-3p targets CAMK2A and regulates the CAMK2A-BDNF pathway in microglia. (A–C) Immunoblot and densitometry analysis shows that overexpression of mir-142-3p mimics suppresses the protein expression levels of CAMK2A and BDNF in BV2 microglia significantly. However, overexpression of miR-142-5p did not suppress CAMK2A protein expression levels. Data represented as mean ± SD, (n = 3), Students t-test, **p < 0.01; *p < 0.05. (D–F) Immunoblot and densitometry quantification shows that overexpression of mir-142-3p inhibitors in BV2 microglia leads to an increase in the protein expression levels of CAMK2A and BDNF, suggesting an inverse relationship between miR-142-3p and CAMK2A. Immunoblot data has been quantified and normalized to β-actin. Data represented as mean ± SD, (n = 3), Students t-test, *p < 0.05. (G) The histogram shows a significant decrease in the luciferase activity in BV2 cells co-transfected with miR-142-3p mimics and luciferase vector as compared to cells co-transfected with negative control miRNA and luciferase vector, indicating that miR-142-3p targets CAMK2A. Data represented as mean ± SD, (n = 3), Students t-test, **p < 0.01.
Figure 6
Figure 6
shRNA mediated knockdown of CAMK2A suppresses the CAMK2A-BDNF pathway in microglia. (A) Quantitative RT-PCR results show a significant decrease in the mRNA levels of Camk2a in BV2 microglia following the shRNA-mediated knockdown of CAMK2A (shCAMK2A). Data represented as mean ± SD, (n = 4), Students t-test, ***p < 0.001. (B) Immunoblot shows that the shRNA mediated knockdown of CAMK2A leads to the downregulation of BDNF via the downregulation of pCREB in BV2 microglia when compared to negative control plasmid (pLKO) transfected microglial cells. (C) The histogram shows the significant downregulation of CAMK2A, pCREB, and BDNF in shCAMK2A microglia, compared to negative control plasmids (pLKO) transfected microglial cells. Immunoblot data has been quantified and normalized to β-actin. Phospho-CREB was then normalized to total CREB. Data represented as mean ± SD, (n = 3), Students t-test, ***p < 0.001; **p < 0.01; *p < 0.05. (D) Immunofluorescence analysis shows that CAMK2A (red) expression appears to be decreased in shCAMK2A microglial cells compared to negative control plasmids (pLKO) transfected microglial cells. (E) Immunofluorescence analysis shows that BDNF (red) expression appears to be decreased in shCAMK2A microglial cells compared to negative control plasmids (pLKO) transfected microglial cells. Lectin (green) used as microglial markers. Nuclei are stained with DAPI (blue), (n = 3), Scale bars = 20 μm.
Figure 7
Figure 7
miR-142-3p epigenetically regulates the CAMK2A-BDNF pathway in microglia. In the activated microglia, miR-142-3p was upregulated, leading to suppression of its target, CAMK2A protein expression, which eventually downregulates the level of BDNF through decreased pCREB (red arrow indicates upregulation and green arrow indicates downregulation). This illustration suggests that under pathological conditions, the CAMK2A-BDNF pathway which is known to be involved in learning and memory is epigenetically regulated in activated microglia.

References

    1. Andreakos E., Sacre S. M., Smith C., Lundberg A., Kiriakidis S., Stonehouse T., et al. . (2004). Distinct pathways of LPS-induced NF-κ B activation and cytokine production in human myeloid and nonmyeloid cells defined by selective utilization of MyD88 and Mal/TIRAP. Blood 103, 2229–2237. 10.1182/blood-2003-04-1356
    1. Ashraf U., Zhu B., Ye J., Wan S., Nie Y., Chen Z., et al. . (2016). MicroRNA-19b-3p modulates Japanese encephalitis virus-mediated inflammation via targeting RNF11. J. Virol. 90, 4780–4795. 10.1128/JVI.02586-15
    1. Baghel M. S., Singh B., Dhuriya Y., Shukla R., Nisha P., Khanna V., et al. . (2018). Postnatal exposure to poly (I:C) impairs learning and memory through changes in synaptic plasticity gene expression in developing rat brain. Neurobiol. Learn. Mem. 155, 379–389. 10.1016/j.nlm.2018.09.005
    1. Banati R. B., Gehrmann J., Schubert P., Kreutzberg G. W. (1993). Cytotoxicity of microglia. Glia 7, 111–118. 10.1002/glia.440070117
    1. Blasi E., Barluzzi R., Bocchini V., Mazzolla R., Bistoni F. (1990). Immortalization of murine microglial cells by a v-raf/v-myc carrying retrovirus. J. Neuroimmunol. 27, 229–237. 10.1016/0165-5728(90)90073-v
    1. Chao C., Hu S., Molitor T., Shaskan E., Peterson P. (1992). Activated microglia mediate neuronal cell injury via a nitric oxide mechanism. J. Immunol. 149, 2736–2741.
    1. Chaudhuri A. D., Yelamanchili S. V., Marcondes M. C. G., Fox H. S. (2013). Up-regulation of microRNA-142 in simian immunodeficiency virus encephalitis leads to repression of sirtuin1. FASEB J. 27, 3720–3729. 10.1096/fj.13-232678
    1. Cook S. G., Bourke A. M., O’Leary H., Zaegel V., Lasda E., Mize-Berge J., et al. . (2018). Analysis of the CaMKIIα and β splice-variant distribution among brain regions reveals isoform-specific differences in holoenzyme formation. Sci. Rep. 8:5448. 10.1038/s41598-018-23779-4
    1. Coultrap S. J., Bayer K. U. (2012). CaMKII regulation in information processing and storage. Trends Neurosci. 35, 607–618. 10.1016/j.tins.2012.05.003
    1. Dheen S. T., Jun Y., Yan Z., Tay S. S., Ling E. A. (2005). Retinoic acid inhibits expression of TNF-α and iNOS in activated rat microglia. Glia 50, 21–31. 10.1002/glia.20153
    1. Fetler L., Amigorena S. (2005). Brain under surveillance: the microglia patrol. Science 309, 392–393. 10.1126/science.1114852
    1. Fritz G., Kaina B. (2001). Ras-related GTPase RhoB represses NF-κB signaling. J. Biol. Chem. 276, 3115–3122. 10.1074/jbc.m005058200
    1. Griffin R., Nally R., Nolan Y., McCartney Y., Linden J., Lynch M. A. (2006). The age-related attenuation in long-term potentiation is associated with microglial activation. J. Neurochem. 99, 1263–1272. 10.1111/j.1471-4159.2006.04165.x
    1. Guedes J., Cardoso A., Pedroso de Lima M. (2013). Involvement of microRNA in microglia-mediated immune response. Clin. Dev. Immunol. 2013:186872. 10.1155/2013/186872
    1. Guo Y., Hong W., Wang X., Zhang P., Körner H., Tu J., et al. . (2019). MicroRNAs in microglia: how do MicroRNAs affect activation, inflammation, polarization of microglia and mediate the interaction between microglia and glioma? Front. Mol. Neurosci. 12:125. 10.3389/fnmol.2019.00125
    1. Gupta N., Shyamasundar S., Patnala R., Karthikeyan A., Arumugam T. V., Ling E.-A., et al. . (2018). Recent progress in therapeutic strategies for microglia-mediated neuroinflammation in neuropathologies. Expert Opin. Ther. Targets 22, 765–781. 10.1080/14728222.2018.1515917
    1. Harada T., Harada C., Kohsaka S., Wada E., Yoshida K., Ohno S., et al. . (2002). Microglia-Müller glia cell interactions control neurotrophic factor production during light-induced retinal degeneration. J. Neurosci. 22, 9228–9236. 10.1523/JNEUROSCI.22-21-09228.2002
    1. Hoshiko M., Arnoux I., Avignone E., Yamamoto N., Audinat E. (2012). Deficiency of the microglial receptor CX3CR1 impairs postnatal functional development of thalamocortical synapses in the barrel cortex. J. Neurosci. 32, 15106–15111. 10.1523/JNEUROSCI.1167-12.2012
    1. Hudmon A., Schulman H. (2002). Neuronal CA2+/calmodulin-dependent protein kinase II: the role of structure and autoregulation in cellular function. Annu. Rev. Biochem. 71, 473–510. 10.1146/annurev.biochem.71.110601.135410
    1. Jadhav S. P., Kamath S. P., Choolani M., Lu J., Dheen S. T. (2014). microRNA-200b modulates microglia-mediated neuroinflammation via the cJun/MAPK pathway. J. Neurochem. 130, 388–401. 10.1111/jnc.12731
    1. Kaminska B., Mota M., Pizzi M. (2016). Signal transduction and epigenetic mechanisms in the control of microglia activation during neuroinflammation. Biochim. Biophys. Acta 1862, 339–351. 10.1016/j.bbadis.2015.10.026
    1. Karthikeyan A., Gupta N., Tang C., Mallilankaraman K., Silambarasan M., Shi M., et al. . (2018). Microglial SMAD4 regulated by microRNA-146a promotes migration of microglia which support tumor progression in a glioma environment. Oncotarget 9, 24950–24969. 10.18632/oncotarget.25116
    1. Karthikeyan A., Patnala R., Jadhav S. P., Eng-Ang L., Thameem Dheen S. (2016). MicroRNAs: key players in microglia and astrocyte mediated inflammation in CNS pathologies. Curr. Med. Chem. 23, 3528–3546. 10.2174/0929867323666160814001040
    1. Kawakubo H., Carey J. L., Brachtel E., Gupta V., Green J. E., Walden P. D., et al. . (2004). Expression of the NF-κB-responsive gene BTG2 is aberrantly regulated in breast cancer. Oncogene 23, 8310–8319. 10.1038/sj.onc.1208008
    1. Kettenmann H., Kirchhoff F., Verkhratsky A. (2013). Microglia: new roles for the synaptic stripper. Neuron 77, 10–18. 10.1016/j.neuron.2012.12.023
    1. Lai S.-W., Chen J.-H., Lin H.-Y., Liu Y.-S., Tsai C.-F., Chang P.-C., et al. . (2018). Regulatory effects of neuroinflammatory responses through brain-derived neurotrophic factor signaling in microglial cells. Mol. Neurobiol. 55, 7487–7499. 10.1007/s12035-018-0933-z
    1. Li Q., Barres B. A. (2018). Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 18, 225–242. 10.1038/nri.2017.125
    1. Li B., Dasgupta C., Huang L., Meng X., Zhang L. (2019). MiRNA-210 induces microglial activation and regulates microglia-mediated neuroinflammation in neonatal hypoxic-ischemic encephalopathy. Cell. Mol. Immunol. [Epub ahead of print]. 10.1038/s41423-019-0257-6
    1. Liu X. B., Murray K. D. (2012). Neuronal excitability and calcium/calmodulin dependent protein kinase type II: location, location, location. Epilepsia 53, 45–52. 10.1111/j.1528-1167.2012.03474.x
    1. Lu D.-Y., Tang C.-H., Yeh W.-L., Wong K.-L., Lin C.-P., Chen Y.-H., et al. . (2009). SDF-1α up-regulates interleukin-6 through CXCR4, PI3K/Akt, ERK, and NF-kappaB-dependent pathway in microglia. Eur. J. Pharmacol. 613, 146–154. 10.1016/j.ejphar.2009.03.001
    1. Malenka R. C., Bear M. F. (2004). LTP and LTD: an embarrassment of riches. Neuron 44, 5–21. 10.1016/j.neuron.2004.09.012
    1. Mattick J. S. (2009). The genetic signatures of noncoding RNAs. PLoS Genet. 5:e1000459. 10.1371/journal.pgen.1000459
    1. Medzhitov R., Janeway C., Jr. (2000). The Toll receptor family and microbial recognition. Trends Microbiol. 8, 452–456. 10.1016/s0966-842x(00)01845-x
    1. Minichiello L. (2009). TrkB signalling pathways in LTP and learning. Nat. Rev. Neurosci. 10, 850–860. 10.1038/nrn2738
    1. Mishra R., Chhatbar C., Singh S. K. (2012). HIV-1 Tat C-mediated regulation of tumor necrosis factor receptor-associated factor-3 by microRNA 32 in human microglia. J. Neuroinflammation 9:131. 10.1186/1742-2094-9-131
    1. Moraes C. A., Santos G., de Sampaio e Spohr T. C., D’Avila J. C., Lima F. R. S., Benjamim C. F., et al. . (2015). Activated microglia-induced deficits in excitatory synapses through IL-1β: implications for cognitive impairment in sepsis. Mol. Neurobiol. 52, 653–663. 10.1007/s12035-014-8868-5
    1. Ni J., Wang X., Chen S., Liu H., Wang Y., Xu X., et al. . (2015). MicroRNA let-7c-5p protects against cerebral ischemia injury via mechanisms involving the inhibition of microglia activation. Brain Behav. Immun. 49, 75–85. 10.1016/j.bbi.2015.04.014
    1. Oshiumi H., Matsuo A., Matsumoto M., Seya T. (2008). Pan-vertebrate toll-like receptors during evolution. Curr. Genomics 9, 488–493. 10.2174/138920208786241234
    1. Parkhurst C. N., Yang G., Ninan I., Savas J. N., Yates J. R., Lafaille J. J., et al. . (2013). Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609. 10.1016/j.cell.2013.11.030
    1. Ponomarev E. D., Veremeyko T., Barteneva N., Krichevsky A. M., Weiner H. L. (2011). MicroRNA-124 promotes microglia quiescence and suppresses EAE by deactivating macrophages via the C/EBP-α-PU.1 pathway. Nat. Med. 17, 64–70. 10.1038/nm.2266
    1. Rastogi M., Srivastava N., Singh S. K. (2018). Exploitation of microRNAs by Japanese Encephalitis virus in human microglial cells. J. Med. Virol. 90, 648–654. 10.1002/jmv.24995
    1. Ritchie M. E., Silver J., Oshlack A., Holmes M., Diyagama D., Holloway A., et al. . (2007). A comparison of background correction methods for two-colour microarrays. Bioinformatics 23, 2700–2707. 10.1093/bioinformatics/btm412
    1. Rodriguez P. L., Sahay S., Olabisi O. O., Whitehead I. P. (2007). ROCK I-mediated activation of NF-κB by RhoB. Cell. Signal. 19, 2361–2369. 10.1016/j.cellsig.2007.07.021
    1. Saba R., Gushue S., Huzarewich R. L., Manguiat K., Medina S., Robertson C., et al. . (2012). MicroRNA 146a (miR-146a) is over-expressed during prion disease and modulates the innate immune response and the microglial activation state. PLoS One 7:e30832. 10.1371/journal.pone.0030832
    1. Saika R., Sakuma H., Noto D., Yamaguchi S., Yamamura T., Miyake S. (2017). MicroRNA-101a regulates microglial morphology and inflammation. J. Neuroinflammation 14:109. 10.1186/s12974-017-0884-8
    1. Saponaro C., Cianciulli A., Calvello R., Dragone T., Iacobazzi F., Panaro M. A. (2012). The PI3K/Akt pathway is required for LPS activation of microglial cells. Immunopharm. Immunot. 34, 858–865. 10.3109/08923973.2012.665461
    1. Saw G., Krishna K., Gupta N., Soong T. W., Mallilankaraman K., Sajikumar S., et al. . (2020). Epigenetic regulation of microglial phosphatidylinositol 3-kinase pathway involved in long-term potentiation and synaptic plasticity in rats. Glia 68, 656–669. 10.1002/glia.23748
    1. Schafer D. P., Lehrman E. K., Stevens B. (2013). The “quad-partite” synapse: microglia-synapse interactions in the developing and mature CNS. Glia 61, 24–36. 10.1002/glia.22389
    1. Schafer D. P., Lehrman E. K., Kautzman A. G., Koyama R., Mardinly A. R., Yamasaki R., et al. . (2012). Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705. 10.1016/j.neuron.2012.03.026
    1. Schafer D. P., Stevens B. (2013). Phagocytic glial cells: sculpting synaptic circuits in the developing nervous system. Curr. Opin. Neurobiol. 23, 1034–1040. 10.1016/j.conb.2013.09.012
    1. Selvamani A., Sathyan P., Miranda R. C., Sohrabji F. (2012). An antagomir to microRNA Let7f promotes neuroprotection in an ischemic stroke model. PLoS One 7:e32662. 10.1371/journal.pone.0032662
    1. Shih R.-H., Wang C.-Y., Yang C.-M. (2015). NF-kappaB signaling pathways in neurological inflammation: a mini review. Front. Mol. Neurosci. 8:77. 10.3389/fnmol.2015.00077
    1. Sierra A., Abiega O., Shahraz A., Neumann H. (2013). Janus-faced microglia: beneficial and detrimental consequences of microglial phagocytosis. Front. Cell. Neurosci. 7:6. 10.3389/fncel.2013.00006
    1. Slota J. A., Booth S. A. (2019). MicroRNAs in neuroinflammation: implications in disease pathogenesis, biomarker discovery and therapeutic applications. Noncoding RNA 5:E35. 10.3390/ncrna5020035
    1. Stephenson J. R., Wang X., Perfitt T. L., Parrish W. P., Shonesy B. C., Marks C. R., et al. . (2017). A novel human CAMK2A mutation disrupts dendritic morphology and synaptic transmission, and causes ASD-related behaviors. J. Neurosci. 37, 2216–2233. 10.1523/JNEUROSCI.2068-16.2017
    1. Su W., Aloi M. S., Garden G. A. (2016). MicroRNAs mediating CNS inflammation: small regulators with powerful potential. Brain Behav. Immun. 52, 1–8. 10.1016/j.bbi.2015.07.003
    1. Sun G., Li H., Rossi J. J. (2009). Sequence context outside the target region influences the effectiveness of miR-223 target sites in the RhoB 3′ UTR. Nucleic Acids Res. 38, 239–252. 10.1093/nar/gkp870
    1. Swulius M. T., Waxham M. (2008). Ca2+/calmodulin-dependent protein kinases. Cell. Mol. Life Sci. 65, 2637–2657. 10.1007/s00018-008-8086-2
    1. Taft R. J., Pang K. C., Mercer T. R., Dinger M., Mattick J. S. (2010). Non-coding RNAs: regulators of disease. J. Pathol. 220, 126–139. 10.1002/path.2638
    1. Talebi F., Ghorbani S., Chan W. F., Boghozian R., Masoumi F., Ghasemi S., et al. . (2017). MicroRNA-142 regulates inflammation and T cell differentiation in an animal model of multiple sclerosis. J. Neuroinflammation 14:55. 10.1186/s12974-017-0832-7
    1. Tao X., Finkbeiner S., Arnold D. B., Shaywitz A. J., Greenberg M. E. (1998). Ca2+ influx regulates BDNF transcription by a CREB family transcription factor-dependent mechanism. Neuron 20, 709–726. 10.1016/s0896-6273(00)81010-7
    1. Thameem Dheen S., Kaur C., Ling E.-A. (2007). Microglial activation and its implications in the brain diseases. Curr. Med. Chem. 14, 1189–1197. 10.2174/092986707780597961
    1. Thounaojam M. C., Kaushik D. K., Kundu K., Basu A. (2014). MicroRNA-29b modulates Japanese encephalitis virus-induced microglia activation by targeting tumor necrosis factor α-induced protein 3. J. Neurochem. 129, 143–154. 10.1111/jnc.12609
    1. Wallet C., De Rovere M., Van Assche J., Daouad F., De Wit S., Gautier V., et al. . (2019). Microglial cells: the main HIV-1 reservoir in the brain. Front. Cell. Infect. Microbiol. 9:362. 10.3389/fcimb.2019.00362
    1. Wang X.-H., Wang T.-L. (2018). MicroRNAs of microglia: wrestling with central nervous system disease. Neural Regen. Res. 13, 2067–2072. 10.4103/1673-5374.241444
    1. Witham J., Ouboussad L., Lefevre P. F. (2013). A NF-κB-dependent dual promoter-enhancer initiates the lipopolysaccharide-mediated transcriptional activation of the chicken lysozyme in macrophages. PLoS One 8:e59389. 10.1371/journal.pone.0059389
    1. Wu Y., Dissing-Olesen L., MacVicar B. A., Stevens B. (2015). Microglia: dynamic mediators of synapse development and plasticity. Trends Immunol. 36, 605–613. 10.1016/j.it.2015.08.008
    1. Xie N., Cui H., Banerjee S., Tan Z., Salomao R., Fu M., et al. . (2014). miR-27a regulates inflammatory response of macrophages by targeting IL-10. J. Immunol. 193, 327–334. 10.4049/jimmunol.1400203
    1. Yan X., Liu J., Ye Z., Huang J., He F., Xiao W., et al. . (2016). CaMKII-mediated CREB phosphorylation is involved in Ca2+-induced BDNF mRNA transcription and neurite outgrowth promoted by electrical stimulation. PLoS One 11:e0162784. 10.1371/journal.pone.0162784
    1. Zhang L., Dong L. Y., Li Y. J., Hong Z., Wei W. S. (2012). miR-21 represses FasL in microglia and protects against microglia-mediated neuronal cell death following hypoxia/ischemia. Glia 60, 1888–1895. 10.1002/glia.22404
    1. Zhang Y., Mou J., Cao L., Zhen S., Huang H., Bao H. (2018). MicroRNA-142–3p relieves neuropathic pain by targeting high mobility group box 1. Int. J. Mol. Med. 41, 501–510. 10.3892/ijmm.2017.3222
    1. Zhao H., Wang J., Gao L., Wang R., Liu X., Gao Z., et al. . (2013). MiRNA-424 protects against permanent focal cerebral ischemia injury in mice involving suppressing microglia activation. Stroke 44, 1706–1713. 10.1161/strokeaha.111.000504
    1. Zhao J., Yue D., Zhou Y., Jia L., Wang H., Guo M., et al. . (2017). The role of MicroRNAs in Aβ deposition and tau phosphorylation in Alzheimer’s disease. Front. Neurol. 8:342. 10.3389/fneur.2017.00342
    1. Zhu Q.-Y., Liu Q., Chen J.-X., Lan K., Ge B.-X. (2010). MicroRNA-101 targets MAPK phosphatase-1 to regulate the activation of MAPKs in macrophages. J. Immunol. 185, 7435–7442. 10.4049/jimmunol.1000798
    1. Zielasek J., Hartung H.-P. (1996). Molecular mechanisms of microglial activation. Adv. Neuroimmunol. 6, 191–222. 10.1016/0960-5428(96)00017-4

Source: PubMed

3
Abonner