Epigenetics and Communication Mechanisms in Microglia Activation with a View on Technological Approaches
Abstract
:1. Role of Microglia in CNS Physiology and Pathology
2. Neuron–Microglia Crosstalk in CNS Physiology and Pathology
2.1. Cytokines
2.2. Chemokines
Fractalkine (CX3CL1)
2.3. Immunoglobulins Superfamily (IgSF)
2.3.1. TREM2
2.3.2. CD200
2.4. Role of Extracellular Vesicles in Neuron–Microglia Crosstalk
2.4.1. Proteins
2.4.2. Lipids
2.4.3. Nucleic Acids
3. Epigenetic Mechanisms Involved in Microglia Activation
3.1. DNA Methylation
3.2. Histones Post-Translational Modifications
3.3. MicroRNAs (miRNAs)
4. Neuronal-Mediated Epigenetic Reprogramming of Microglia in CNS Health and Disease
5. Studying Microglia–Neuron Crosstalk with Advanced Microscopy Techniques
6. 3D-Cell Culture Systems as Models for The Interaction of Microglia and Neurons
7. Microfluidics Technologies for Microglia–Neuron Interaction Studies
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
Abbreviations
References
- Kettenmann, H.; Hanisch, U.K.; Noda, M.; Verkhratsky, A. Physiology of microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef] [PubMed]
- Cuadros, M.A.; Martin, C.; Coltey, P.; Almendros, A.; Navascués, J. First appearance, distribution, and origin of macrophages in the early development of the avian central nervous system. J. Comp. Neurol. 1993, 330, 113–129. [Google Scholar] [CrossRef] [PubMed]
- Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef] [Green Version]
- Xu, Y.; Xu, Y.; Wang, Y.; Wang, Y.; He, L.; Jiang, Z.; Huang, Z.; Liao, H.; Li, J.; Saavedra, J.M.; et al. Telmisartan prevention of LPS-induced microglia activation involves M2 microglia polarization via CaMKKβ-dependent AMPK activation. Brain Behav. Immun. 2015, 50, 298–313. [Google Scholar] [CrossRef] [PubMed]
- Lisi, L.; Ciotti, G.M.; Braun, D.; Kalinin, S.; Currò, D.; Russo, C.D.; Coli, A.; Mangiola, A.; Anile, C.; Feinstein, D.L.; et al. Expression of iNOS, CD163 and ARG-1 taken as M1 and M2 markers of microglial polarization in human glioblastoma and the surrounding normal parenchyma. Neurosci. Lett. 2017, 645, 106–112. [Google Scholar] [CrossRef]
- Meng, X.; Duan, C.; Pang, H.; Chen, Q.; Han, B.; Zha, C.; Dinislam, M.; Wu, P.; Li, Z.; Zhao, S.; et al. DNA damage repair alterations modulate M2 polarization of microglia to remodel the tumor microenvironment via the p53-mediated MDK expression in glioma. EBioMedicine 2019, 41, 185–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chamera, K.; Trojan, E.; Szuster-Głuszczak, M.; Basta-Kaim, A. The Potential Role of Dysfunctions in Neuron-Microglia Communication in the Pathogenesis of Brain Disorders. Curr. Neuropharmacol. 2020, 18, 408–430. [Google Scholar] [CrossRef]
- Wake, H.; Moorhouse, A.J.; Nabekura, J. Functions of microglia in the central nervous system--beyond the immune response. Neuron Glia Biol. 2011, 7, 47–53. [Google Scholar] [CrossRef] [PubMed]
- Eyo, U.B.; Wu, L.J. Bidirectional microglia-neuron communication in the healthy brain. Neural Plast. 2013, 2013, 456857. [Google Scholar] [CrossRef]
- Marinelli, S.; Basilico, B.; Marrone, M.C.; Ragozzino, D. Microglia-neuron crosstalk: Signaling mechanism and control of synaptic transmission. Semin. Cell Dev. Biol. 2019, 94, 138–151. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Xu, J.; Gao, J.; Wu, Y.; Yin, M.; Zhao, W. CD200-, CX3CL1-, and TREM2-mediated neuron-microglia interactions and their involvements in Alzheimer’s disease. Rev. Neurosci. 2018, 29, 837–848. [Google Scholar] [CrossRef]
- Sato, K. Effects of Microglia on Neurogenesis. Glia 2015, 63, 1394–1405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Šišková, Z.; Tremblay, M.È. Microglia and synapse: Interactions in health and neurodegeneration. Neural Plast. 2013, 2013, 425845. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.; Dissing-Olesen, L.; MacVicar, B.A.; Stevens, B. Microglia: Dynamic Mediators of Synapse Development and Plasticity. Trends Immunol. 2015, 36, 605–613. [Google Scholar] [CrossRef] [Green Version]
- Wake, H.; Moorhouse, A.J.; Miyamoto, A.; Nabekura, J. Microglia: Actively surveying and shaping neuronal circuit structure and function. Trends Neurosci. 2013, 36, 209–217. [Google Scholar] [CrossRef] [PubMed]
- Deverman, B.E.; Patterson, P.H. Cytokines and CNS development. Neuron 2009, 64, 61–78. [Google Scholar] [CrossRef] [Green Version]
- Kerschensteiner, M.; Meinl, E.; Hohlfeld, R. Neuro-immune crosstalk in CNS diseases. Neuroscience 2009, 158, 1122–1132. [Google Scholar] [CrossRef]
- Smith, J.A.; Das, A.; Ray, S.K.; Banik, N.L. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res. Bull. 2012, 87, 10–20. [Google Scholar] [CrossRef]
- Biber, K.; Neumann, H.; Inoue, K.; Boddeke, H.W. Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci. 2007, 30, 596–602. [Google Scholar] [CrossRef]
- Hanisch, U.K. Microglia as a source and target of cytokines. Glia 2002, 40, 140–155. [Google Scholar] [CrossRef]
- Fixe, P.; Praloran, V. M-CSF: Haematopoietic growth factor or inflammatory cytokine? Cytokine 1998, 10, 32–37. [Google Scholar] [CrossRef]
- Wyss-Coray, T.; Lin, C.; Yan, F.; Yu, G.Q.; Rohde, M.; McConlogue, L.; Masliah, E.; Mucke, L. TGF-beta1 promotes microglial amyloid beta clearance and reduces plaque burden in transgenic mice. Nat. Med. 2001, 7, 612–618. [Google Scholar] [CrossRef]
- Suzumura, A. Microglia in neurodegenerative disorders and neuroinflammation. Rinsho Shinkeigaku 2014, 54, 1119–1121. [Google Scholar] [CrossRef] [PubMed]
- Akiyama, H.; Barger, S.; Barnum, S.; Bradt, B.; Bauer, J.; Cole, G.M.; Cooper, N.R.; Eikelenboom, P.; Emmerling, M.; Fiebich, B.L.; et al. Inflammation and Alzheimer’s disease. Neurobiol. Aging 2000, 21, 383–421. [Google Scholar] [CrossRef]
- McGeer, P.L.; McGeer, E.G. Inflammation, autotoxicity and Alzheimer disease. Neurobiol. Aging 2001, 22, 799–809. [Google Scholar] [CrossRef]
- Parajuli, B.; Horiuchi, H.; Mizuno, T.; Takeuchi, H.; Suzumura, A. CCL11 enhances excitotoxic neuronal death by producing reactive oxygen species in microglia. Glia 2015, 63, 2274–2284. [Google Scholar] [CrossRef] [PubMed]
- Feng, C.; Wang, X.; Liu, T.; Zhang, M.; Xu, G.; Ni, Y. Expression of CCL2 and its receptor in activation and migration of microglia and monocytes induced by photoreceptor apoptosis. Mol. Vis. 2017, 23, 765–777. [Google Scholar]
- Ransohoff, R.M.; El Khoury, J. Microglia in Health and Disease. Cold Spring Harb. Perspect. Biol. 2015, 8, a020560. [Google Scholar] [CrossRef] [Green Version]
- Paolicelli, R.C.; Bisht, K.; Tremblay, M.E. Fractalkine regulation of microglial physiology and consequences on the brain and behavior. Front. Cell Neurosci. 2014, 8, 129. [Google Scholar] [CrossRef] [Green Version]
- Zujovic, V.; Benavides, J.; Vige, X.; Carter, C.; Taupin, V. Fractalkine modulates TNF-alpha secretion and neurotoxicity induced by microglial activation. Glia 2000, 29, 305–315. [Google Scholar] [CrossRef]
- Gemma, C.; Bachstetter, A.D.; Bickford, P.C. Neuron-Microglia Dialogue and Hippocampal Neurogenesis in the Aged Brain. Aging Dis. 2010, 1, 232–244. [Google Scholar] [PubMed]
- Rogers, J.T.; Morganti, J.M.; Bachstetter, A.D.; Hudson, C.E.; Peters, M.M.; Grimmig, B.A.; Weeber, E.J.; Bickford, P.C.; Gemma, C. CX3CR1 deficiency leads to impairment of hippocampal cognitive function and synaptic plasticity. J. Neurosci. 2011, 31, 16241–16250. [Google Scholar] [CrossRef] [Green Version]
- Cardona, A.E. Control of microglial neurotoxicity by the fractalkine receptor. Nat. Neurosci. 2006, 9, 917–924. [Google Scholar] [CrossRef]
- Uyemura, K.; Asou, H.; Yazaki, T.; Takeda, Y. Cell-adhesion proteins of the immunoglobulin superfamily in the nervous system. Essays Biochem. 1996, 31, 37–48. [Google Scholar] [PubMed]
- Jiang, T.; Tan, L.; Zhu, X.C.; Zhang, Q.Q.; Cao, L.; Tan, M.S.; Gus, L.Z.; Wang, H.F.; Ding, Z.Z.; Zhang, Y.D.; et al. Upregulation of TREM2 ameliorates neuropathology and rescues spatial cognitive impairment in a transgenic mouse model of Alzheimer’s disease. Neuropsychopharmacology 2014, 39, 2949–2962. [Google Scholar] [CrossRef] [Green Version]
- Painter, M.M.; Atagi, Y.; Liu, C.C.; Rademakers, R.; Xu, H.; Fryer, J.D.; Bu, G. TREM2 in CNS homeostasis and neurodegenerative disease. Mol. Neurodegener. 2015, 10, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Edmonson, C.; Ziats, M.N.; Rennert, O.M. Altered glial marker expression in autistic post-mortem prefrontal cortex and cerebellum. Mol. Autism. 2014, 10, 3. [Google Scholar] [CrossRef] [Green Version]
- Filipello, F.; Morini, R.; Corradini, I.; Zerbi, V.; Canzi, A.; Michalski, B.; Erreni, M.; Markicevic, M.; Starvaggi-Cucuzza, C.; Otero, K.; et al. The microglial innate immune receptor TREM2 is required for synapse elimination and normal brain connectivity. Immunity 2018, 48, 979–991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walker, D.G.; Lue, L.F. Immune phenotypes of microglia in human neurodegenerative disease: Challenges to detecting microglial polarization in human brains. Alzheimers Res. Ther. 2015, 7, 56. [Google Scholar] [CrossRef] [Green Version]
- Lyons, A.; Downer, E.J.; Crotty, S.; Nolan, Y.M.; Mills, K.H.G.; Lynch, M.A. CD200 ligand-receptor interaction modulates microglial activation in vivo and in vitro: A role for IL-4. J. Neurosci. 2007, 27, 8309–8313. [Google Scholar] [CrossRef]
- Lyons, A.; McQuillan, K.; Deighan, B.F.; O’Reilly, J.A.; Downer, E.J.; Murphy, A.C.; Watson, M.; Piazza, A.; O’Connell, F.; Griffin, R.; et al. Decreased neuronal CD200 expression in IL-4-deficient mice results in increased neuroinflammation in response to lipopolysaccharide. Brain Behav. Immun. 2009, 23, 1020–1027. [Google Scholar] [CrossRef]
- Trotta, T.; Panaro, M.A.; Cianciulli, A.; Mori, G.; Di Benedetto, A.; Porro, C. Microglia-derived extracellular vesicles in Alzheimer’s Disease: A double-edged sword. Biochem. Pharmacol. 2018, 148, 184–192. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
- Losurdo, M.; Pedrazzoli, M.; D’Agostino, C.; Elia, C.A.; Massenzio, F.; Lonati, E.; Mauri, M.; Rizzi, L.; Molteni, L.; Bresciani, E.; et al. Intranasal delivery of mesenchymal stem cell-derived extracellular vesicles exerts immunomodulatory and neuroprotective effects in a 3xTg model of Alzheimer’s disease. Stem Cells Transl. Med. 2020, 9, 1068–1084. [Google Scholar] [CrossRef]
- Budnik, V.; Ruiz-Cañada, C.; Wendler, F. Extracellular vesicles round off communication in the nervous system. Nat. Rev. Neurosci. 2016, 17, 160–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Haas, A.H.; Van Weering, H.R.J.; De Jong, E.K.; Boddeke, H.W.G.M.; Biber, K.P.H. Neuronal chemokines: Versatile messengers in central nervous system cell interaction. Mol. Neurobiol. 2007, 36, 137–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoon, Y.J.; Kim, O.Y.; Gho, Y.S. Extracellular vesicles as emerging intercellular communicasomes. BMB Rep. 2014, 47, 531–539. [Google Scholar] [CrossRef] [PubMed]
- Glebov, K.; Löchner, M.; Jabs, R.; Lau, T.; Merkel, O.; Schloss, P.; Steinhäuser, C.; Walter, J. Serotonin stimulates secretion of exosomes from microglia cells. Glia 2015, 63, 626–634. [Google Scholar] [CrossRef]
- Schneider, A.; Simons, M. Exosomes: Vesicular carriers for intercellular communication in neurodegenerative disorders. Cell Tissue Res. 2013, 352, 33–47. [Google Scholar] [CrossRef] [Green Version]
- Vingtdeux, V.; Sergeant, N.; Buee, L. Potential contribution of exosomes to the prion-like propagation of lesions in Alzheimer’s disease. Front. Physiol. 2012, 3, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antonucci, F.; Turola, E.; Riganti, L.; Caleo, M.; Gabrielli, M.; Perrotta, C.; Novellino, L.; Clementi, E.; Giussani, P.; Viani, P.; et al. Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO J. 2012, 7, 1231–1240. [Google Scholar] [CrossRef] [PubMed]
- Gabrielli, M.; Battista, N.; Riganti, L.; Prada, I.; Antonucci, F.; Cantone, L.; Matteoli, M.; Maccarrone, M.; Verderio, C. Active endocannabinoids are secreted on extracellular membrane vesicles. EMBO Rep. 2015, 16, 213–220. [Google Scholar] [CrossRef] [Green Version]
- Kalluri, R.; LeBleu, V.S. Discovery of Double-Stranded Genomic DNA in Circulating Exosomes. Cold Spring Harb. Symp. Quant. Biol. 2016, 81, 275–280. [Google Scholar] [CrossRef] [Green Version]
- Etheridge, A.; Lee, I.; Hood, L.; Galas, D.; Wang, K. Extracellular microRNA: A new source of biomarkers. Mutat. Res. 2011, 717, 85–90. [Google Scholar] [CrossRef] [Green Version]
- Alexander, M.; Hu, R.; Runtsch, M.C.; Kagele, D.A.; Mosbruger, T.L.; Tolmachova, T.; Seabra, M.C.; Round, J.L.; Ward, D.M.; O’Connell, R.M. Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Nat. Commun. 2015, 18, 7321. [Google Scholar] [CrossRef] [Green Version]
- Lemaire, Q.; Raffo-Romero, A.; Arab, T.; Van Camp, C.; Drago, F.; Forte, S.; Gimeno, J.P.; Begard, S.; Colin, M.; Vizioli, J.; et al. Isolation of microglia-derived extracellular vesicles: Towards miRNA signatures and neuroprotection. J. Nanobio Technol. 2019, 17, 119. [Google Scholar] [CrossRef] [PubMed]
- Prada, I.; Gabrielli, M.; Turola, E.; Iorio, A.; D’Arrigo, G.; Parolisi, R.; De Luca, M.; Pacifici, M.; Bastoni, M.; Lombardi, M.; et al. Glia-to-neuron transfer of miRNAs via extracellular vesicles: A new mechanism underlying inflammation-induced synaptic alterations. Acta Neuropathol. 2018, 135, 529–550. [Google Scholar] [CrossRef] [Green Version]
- Morton, M.C.; Neckles, V.N.; Seluzicki, C.M.; Holmberg, J.C.; Feliciano, D.M. Neonatal Subventricular Zone Neural Stem Cells Release Extracellular Vesicles that Act as a Microglial Morphogen. Cell Rep. 2018, 23, 78–89. [Google Scholar] [CrossRef] [Green Version]
- Pinto, S.; Cunha, C.; Barbosa, M.; Vaz, A.R.; Brites, D. Exosomes from NSC-34 Cells Transfected with hSOD1-G93A are Enriched in miR-124 and Drive Alterations in Microglia Phenotype. Front. Neurosci. 2017, 11, 273. [Google Scholar] [CrossRef] [Green Version]
- Song, Y.; Li, Z.; He, T.; Qu, M.; Jiang, L.; Li, W.; Shi, X.; Pan, J.; Zhang, L.; Wang, Y.; et al. M2 microglia-derived exosomes protect the mouse brain from ischemia-reperfusion injury via exosomal miR-124. Theranostics 2019, 9, 2910–2923. [Google Scholar] [CrossRef] [PubMed]
- Sohel, M.H. Extracellular/Circulating MicroRNAs: Release Mechanisms, Functions and Challenges. Achiev. Life Sci. 2016, 10, 175–186. [Google Scholar] [CrossRef] [Green Version]
- Akers, J.C.; Ramakrishnan, V.; Kim, R.; Skog, J.; Nakano, I.; Pingle, S.; Kalinina, J.; Hua, W.; Kesari, S.; Mao, Y.; et al. MiR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): A platform for glioblastoma biomarker development. PLoS ONE 2013, 8, e78115. [Google Scholar] [CrossRef]
- Zhang, L.; Dong, L.Y.; Li, Y.J.; Hong, Z.; Wei, W.S. The microRNA miR-181c controls microglia-mediated neuronal apoptosis by suppressing tumor necrosis factor. J. Neuroinflamm. 2012, 9, 211. [Google Scholar] [CrossRef] [Green Version]
- Vaz, A.R.; Pinto, S.; Ezequiel, C.; Cunha, C.; Carvalho, L.A.; Moreira, R.; Brites, D. Phenotypic Effects of Wild-Type and Mutant SOD1 Expression in N9 Murine Microglia at Steady State, Inflammatory and Immunomodulatory Conditions. Front. Cell Neurosci. 2019, 13, 109. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Xu, C.; Nan, Y.; Nan, S. Microglia-Derived Extracellular Vesicles Carrying miR-711 Alleviate Neurodegeneration in a Murine Alzheimer’s Disease Model by Binding to Itpkb. Front. Cell Dev. Biol. 2020, 8, 566530. [Google Scholar] [CrossRef]
- Chen, O.; Donnelly, C.R.; Ji, R.R. Regulation of pain by neuro-immune interactions between macrophages and nociceptor sensory neurons. Curr. Opin. Neurobiol. 2020, 62, 17–25. [Google Scholar] [CrossRef] [PubMed]
- Fernandes, A.; Ribeiro, A.R.; Monteiro, M.; Garcia, G.; Vaz, A.R.; Brites, D. Secretome from SH-SY5Y APP Swe cells trigger time-dependent CHME3 microglia activation phenotypes, ultimately leading to miR-21 exosome shuttling. Biochimie 2018, 155, 67–82. [Google Scholar] [CrossRef]
- Ge, X.; Guo, M.; Hu, T.; Li, W.; Huang, S.; Yin, Z.; Zhang, J. Increased Microglial Exosomal miR-124-3p Alleviates Neurodegeneration and Improves Cognitive Outcome after rmTBI. Mol. Ther. 2020, 28, 503–522. [Google Scholar] [CrossRef] [Green Version]
- Han, C.; Guo, L.; Yang, Y.; Guan, Q.; Shen, H.; Sheng, Y.; Jiao, Q. Mechanism of microRNA-22 in regulating neuroinflammation in Alzheimer’s disease. Brain Behav. 2020, 10, e01627. [Google Scholar] [CrossRef] [PubMed]
- Esteller, M. Epigenetics in cancer. N. Engl. J. Med. 2008, 358, 1148–1159. [Google Scholar] [CrossRef] [PubMed]
- Bradley-Whitman, M.A.; Lovell, M.A. Epigenetic changes in the progression of Alzheimer’s disease. Mech. Aging Dev. 2013, 134, 486–495. [Google Scholar] [CrossRef] [Green Version]
- Coppieters, N.; Dieriks, B.V.; Lill, C.; Faull, R.L.; Curtis, M.A.; Dragunow, M. Global changes in DNA methylation and hydroxymethylation in Alzheimer’s disease human brain. Neurobiol. Aging 2014, 35, 1334–1344. [Google Scholar] [CrossRef] [PubMed]
- Day, J.J.; Sweatt, J.D. Cognitive neuroepigenetics: A role for epigenetic mechanisms in learning and memory. Neurobiol. Learn Mem. 2011, 96, 2–12. [Google Scholar] [CrossRef] [Green Version]
- Pang, K.K.L.; Sharma, M.; Sajikumar, S. Epigenetics and memory: Emerging role of histone lysine methyltransferase G9a/GLP complex as bidirectional regulator of synaptic plasticity. Neurobiol. Learn Mem. 2019, 159, 1–5. [Google Scholar] [CrossRef]
- Cho, S.H.; Chen, J.A.; Sayed, F.; Ward, M.E.; Gao, F.; Nguyen, T.A.; Krabbe, G.; Sohn, P.D.; Lo, I.; Minami, S.; et al. SIRT1 deficiency in microglia contributes to cognitive decline in aging and neurodegeneration via epigenetic regulation of IL-1beta. J. Neurosci. 2015, 14, 35–807. [Google Scholar] [CrossRef] [Green Version]
- Matt, S.M.; Lawson, M.A.; Johnson, R.W. Aging and peripheral lipopolysaccharide can modulate epigenetic regulators and decrease IL-1β promoter DNA methylation in microglia. Neurobiol. Aging 2016, 47, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bakulski, K.M.; Dolinoy, D.C.; Sartor, M.A.; Paulson, H.L.; Konen, J.R.; Lieberman, A.P.; Albin, R.L.; Hu, H.; Rozek, L.S. Genome-wide DNA methylation differences between late-onset Alzheimer’s disease and cognitively normal controls in human frontal cortex. J. Alzheimers Dis. 2012, 29, 571–588. [Google Scholar] [CrossRef] [Green Version]
- Mastroeni, D.; Grover, A.; Delvaux, E.; Whiteside, C.; Coleman, P.D.; Rogers, J. Epigenetic changes in Alzheimer’s disease: Decrements in DNA methylation. Neurobiol. Aging. 2010, 31, 2025–2037. [Google Scholar] [CrossRef] [Green Version]
- Chouliaras, L.; Mastroeni, D.; Delvaux, E.; Grover, A.; Kenis, G.; Hof, P.R.; Steinbusch, H.W.; Coleman, P.D.; Rutten, B.P.; van den Hove, D.L. Consistent decrease in global DNA methylation and hydroxymethylation in the hippocampus of Alzheimer’s disease patients. Neurobiol. Aging 2013, 34, 2091–2099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sathasivam, K.; Neueder, A.; Gipson, T.A.; Landles, C.; Benjamin, A.C.; Bondulich, M.K.; Smith, D.L.; Faull, R.L.; Roos, R.A.; Howland, D.; et al. Aberrant splicing of HTT generates the pathogenic exon 1 protein in Huntington disease. Proc. Natl. Acad. Sci. USA 2013, 110, 2366–2370. [Google Scholar] [CrossRef] [Green Version]
- Sanchez-Mut, J.V.; Heyn, H.; Vidal, E.; Moran, S.; Sayols, S.; Delgado-Morales, R.; Schultz, M.D.; Ansoleaga, B.; Garcia-Esparcia, P.; Pons-Espinal, M.; et al. Human DNA methylomes of neurodegenerative diseases show common epigenomic patterns. Transl. Psychiatry 2016, 6, e718. [Google Scholar] [CrossRef] [Green Version]
- Gijselinck, I.; Van Mossevelde, S.; van der Zee, J.; Sieben, A.; Engelborghs, S.; De Bleecker, J.; Ivanoiu, A.; Deryck, O.; Edbauer, D.; Zhang, M.; et al. The C9orf72 repeat size correlates with onset age of disease, DNA methylation and transcriptional downregulation of the promoter. Mol. Psychiatry 2016, 21, 1112–1124. [Google Scholar] [CrossRef] [Green Version]
- Lin, H.C.; Hsieh, H.M.; Chen, Y.H.; Hu, M.L. S-Adenosylhomocysteine increases beta-amyloid formation in BV-2 microglial cells by increased expressions of beta-amyloid precursor protein and presenilin 1 and by hypomethylation of these gene promoters. Neurotoxicology 2009, 30, 622–627. [Google Scholar] [CrossRef]
- Byun, C.J.; Seo, J.; Jo, S.A.; Park, Y.J.; Klug, M.; Rehli, M.; Park, M.H.; Jo, I. DNA methylation of the 5′-untranslated region at +298 and +351 represses BACE1 expression in mouse BV-2 microglial cells. Biochem. Biophys. Res. Commun. 2012, 417, 387–392. [Google Scholar] [CrossRef] [PubMed]
- Moussa-Pacha, N.M.; Abdin, S.M.; Omar, H.A.; Alniss, H.; Al-Tel, T.H. BACE1 inhibitors: Current status and future directions in treating Alzheimer’s disease. Med. Res. Rev. 2020, 40, 339–384. [Google Scholar] [CrossRef] [PubMed]
- Prati, F.; De Simone, A.; Armirotti, A.; Summa, M.; Pizzirani, D.; Scarpelli, R.; Bertozzi, S.M.; Perez, D.I.; Andrisano, V.; Perez-Castillo, A.; et al. 3,4-Dihydro-1,3,5-triazin-2(1H)-ones as the First Dual BACE-1/GSK-3β Fragment Hits against Alzheimer’s Disease. ACS Chem. Neurosci. 2015, 6, 1665–1682. [Google Scholar] [CrossRef] [PubMed]
- Griñán-Ferré, C.; Marsal-García, L.; Bellver-Sanchis, A.; Kondengaden, S.M.; Turga, R.C.; Vázquez, S.; Pallàs, M. Pharmacological inhibition of G9a/GLP restores cognition and reduces oxidative stress, neuroinflammation and β-Amyloid plaques in an early-onset Alzheimer’s disease mouse model. Aging 2019, 11, 11591–11608. [Google Scholar] [CrossRef]
- Kelly, R.D.; Cowley, S.M. The physiological roles of histone deacetylase (HDAC) 1 and 2: Complex co-stars with multiple leading parts. Biochem. Soc. Trans. 2013, 41, 741–749. [Google Scholar] [CrossRef] [Green Version]
- Datta, M.; Staszewski, O.; Raschi, E.; Frosch, M.; Hagemeyer, N.; Tay, T.L.; Blank, T.; Kreutzfeldt, M.; Merkler, D.; Ziegler-Waldkirch, S.; et al. Histone deacetylases 1 and 2 regulate microglia function during development, homeostasis, and neurodegeneration in a context-dependent manner. Immunity 2018, 48, 514–529.e6. [Google Scholar] [CrossRef] [Green Version]
- Buonvicino, D.; Felici, R.; Ranieri, G.; Caramelli, R.; Lapucci, A.; Cavone, L.; Muzzi, M.; Di Pietro, L.; Bernardini, C.; Zwergel, C.; et al. Effects of Class II-Selective Histone Deacetylase Inhibitor on Neuromuscular Function and Disease Progression in SOD1-ALS Mice. Neuroscience 2018, 379, 228–238. [Google Scholar] [CrossRef] [PubMed]
- Harrison, I.F.; Crum, W.R.; Vernon, A.C.; Dexter, D.T. Neurorestoration induced by the HDAC inhibitor sodium valproate in the lactacystin model of Parkinson’s is associated with histone acetylation and up-regulation of neurotrophic factors. Br. J. Pharmacol. 2015, 172, 4200–4215. [Google Scholar] [CrossRef] [Green Version]
- Kim, T.; Song, S.; Park, Y.; Kang, S.; Seo, H. HDAC Inhibition by Valproic Acid Induces Neuroprotection and Improvement of PD-like Behaviors in LRRK2 R1441G Transgenic Mice. Exp. Neurobiol. 2019, 28, 504–515. [Google Scholar] [CrossRef] [Green Version]
- Monti, B.; Gatta, V.; Piretti, F.; Raffaelli, S.S.; Virgili, M.; Contestabile, A. Valproic acid is neuroprotective in the rotenone rat model of Parkinson’s disease: Involvement of alpha-synuclein. Neurotox. Res. 2010, 17, 130–141. [Google Scholar] [CrossRef]
- Romeiro, L.A.S.; da Nunes, J.L.C.; de Miranda, C.O.; Cardoso, G.S.H.R.; de Oliveira, A.S.; Gandini, A.; Kobrlova, T.; Soukup, O.; Rossi, M.; Senger, J.; et al. Novel Sustainable-by-Design HDAC Inhibitors for the Treatment of Alzheimer’s Disease. ACS Med. Chem. Lett. 2019, 10, 671–676. [Google Scholar] [CrossRef] [PubMed]
- Han, S.B.; Lee, J.K. Anti-inflammatory effect of Trichostatin-A on murine bone marrow-derived macrophages. Arch. Pharm. Res. 2009, 32, 613–624. [Google Scholar] [CrossRef] [PubMed]
- Kannan, V.; Brouwer, N.; Hanisch, U.K.; Regen, T.; Eggen, B.J.; Boddeke, H.W. Histone deacetylase inhibitors suppress immune activation in primary mouse microglia. J. Neurosci. Res. 2013, 91, 1133–1142. [Google Scholar] [CrossRef]
- Lin, F.L.; Yen, J.L.; Kuo, Y.C.; Kang, J.J.; Cheng, Y.W.; Huang, W.J.; Hsiao, G. HADC8 Inhibitor WK2-16 Therapeutically Targets Lipopolysaccharide-Induced Mouse Model of Neuroinflammation and Microglial Activation. Int. J. Mol. Sci. 2019, 20, 410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.Y.; Fan, S.J.; Huang, F.I.; Chao, H.Y.; Hsu, K.C.; Lin, T.E.; Yeh, T.K.; Lai, M.J.; Li, Y.H.; Huang, H.L.; et al. 5-Aroylindoles Act as Selective Histone Deacetylase 6 Inhibitors Ameliorating Alzheimer’s Disease Phenotypes. J. Med. Chem. 2018, 61, 7087–7102. [Google Scholar] [CrossRef]
- Gal, J.; Chen, J.; Barnett, K.R.; Yang, L.; Brumley, E.; Zhu, H. HDAC6 regulates mutant SOD1 aggregation through two SMIR motifs and tubulin acetylation. J. Biol. Chem. 2013, 288, 15035–15045. [Google Scholar] [CrossRef] [Green Version]
- Taes, I.; Timmers, M.; Hersmus, N.; Bento-Abreu, A.; Van Den Bosch, L.; Van Damme, P.; Robberecht, W. Hdac6 deletion delays disease progression in the SOD1G93A mouse model of ALS. Hum. Mol. Genet. 2013, 22, 1783–1790. [Google Scholar] [CrossRef]
- Guo, W.; Naujock, M.; Fumagalli, L.; Vandoorne, T.; Baatsen, P.; Boon, R.; Ordovás, L.; Patel, A.; Welters, M.; Vanwelden, T.; et al. HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients. Nat. Commun. 2017, 8, 861. [Google Scholar] [CrossRef]
- Iwamoto, M.; Nakamura, Y.; Takemura, M.; Hisaoka-Nakashima, K.; Morioka, N. TLR4-TAK1-p38 MAPK pathway and HDAC6 regulate the expression of sigma-1 receptors in rat primary cultured microglia. J. Pharmacol. Sci. 2020, 144, 23–29. [Google Scholar] [CrossRef] [PubMed]
- Chao, J.; Zhang, Y.; Du, L.; Zhou, R.; Wu, X.; Shen, K.; Yao, H. Molecular mechanisms underlying the involvement of the sigma-1 receptor in methamphetamine-mediated microglial polarization. Sci. Rep. 2017, 7, 11540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiao, F.Z.; Wang, Y.; Zhang, H.Y.; Zhang, W.B.; Wang, L.W.; Gong, Z.J. Histone Deacetylase 2 Inhibitor CAY10683 Alleviates Lipopolysaccharide Induced Neuroinflammation Through Attenuating TLR4/NF-κB Signaling Pathway. Neurochem. Res. 2018, 43, 1161–1170. [Google Scholar] [CrossRef]
- Chen, P.S.; Peng, G.S.; Li, G.; Yang, S.; Wu, X.; Wang, C.C.; Wilson, B.; Lu, R.B.; Gean, P.W.; Chuang, D.M.; et al. Valproate protects dopaminergic neurons in midbrain neuron/glia cultures by stimulating the release of neurotrophic factors from astrocytes. Mol. Psychiatry 2006, 11, 1116–1125. [Google Scholar] [CrossRef] [Green Version]
- Harrison, I.F.; Smith, A.D.; Dexter, D.T. Pathological histone acetylation in Parkinson’s disease: Neuroprotection and inhibition of microglial activation through SIRT 2 inhibition. Neurosci. Lett. 2018, 666, 48–57. [Google Scholar] [CrossRef] [PubMed]
- Peng, G.S.; Li, G.; Tzeng, N.S.; Chen, P.S.; Chuang, D.M.; Hsu, Y.D.; Yang, S.; Hong, J.S. Valproate pretreatment protects dopaminergic neurons from LPS-induced neurotoxicity in rat primary midbrain cultures: Role of microglia. Brain Res. Mol. Brain Res. 2005, 134, 162–169. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.; Zhang, Y.; Gong, Y.; Yang, R.; Chen, Z.; Hu, W.; Wu, Y.; Gao, M.; Xu, X.; Qin, Y.; et al. Sodium butyrate triggers a functional elongation of microglial process via Akt-small RhoGTPase activation and HDACs inhibition. Neurobiol. Dis. 2018, 111, 12–25. [Google Scholar] [CrossRef] [PubMed]
- Slota, J.A.; Booth, S.A. MicroRNAs in Neuroinflammation: Implications in Disease Pathogenesis, Biomarker Discovery and Therapeutic Applications. Noncoding RNA 2019, 5, 35. [Google Scholar] [CrossRef] [Green Version]
- Jonas, S.; Izaurralde, E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat. Rev. Genet. 2015, 16, 421–433. [Google Scholar] [CrossRef]
- Gulyaeva, L.F.; Kushlinskiy, N.E. Regulatory mechanisms of microRNA expression. J. Transl. Med. 2016, 14, 143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paolicelli, R.C.; Bergamini, G.; Rajendran, L. Cell-to-cell Communication by Extracellular Vesicles: Focus on Microglia. Neuroscience 2019, 405, 148–157. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Li, S.; Li, L.; Li, M.; Guo, C.; Yao, J.; Mi, S. Exosome and Exosomal MicroRNA: Trafficking, Sorting, and Function, Genomics, Proteomics. Bioinformatics 2015, 13, 17–24. [Google Scholar] [CrossRef] [Green Version]
- Freilich, R.W.; Woodbury, M.E.; Ikezu, T. Integrated expression profiles of mRNA and miRNA in polarized primary murine microglia. PLoS ONE 2013, 8, e79416. [Google Scholar] [CrossRef] [Green Version]
- Cardoso, A.L.; Guedes, J.R.; de Almeida, L.P.; de Lima, M.C.P. miR-155 modulates microglia-mediated immune response by down-regulating SOCS-1 and promoting cytokine and nitric oxide production. Immunology 2012, 135, 73–88. [Google Scholar] [CrossRef] [PubMed]
- Martin, N.A.; Hyrlov, K.H.; Elkjaer, M.L.; Thygesen, E.K.; Wlodarczyk, A.; Elbaek, K.J.; Aboo, C.; Okarmus, J.; Benedikz, E.; Reynolds, R.; et al. Absence of miRNA-146a Differentially Alters Microglia Function and Proteome. Front. Immunol. 2020, 11, 1110. [Google Scholar] [CrossRef]
- Tahamtan, A.; Teymoori-Rad, M.; Nakstad, B.; Salimi, V. Anti-Inflammatory MicroRNAs and Their Potential for Inflammatory Diseases Treatment. Front. Immunol. 2018, 9, 1377. [Google Scholar] [CrossRef] [Green Version]
- Vergadi, E.; Vaporidi, K.; Theodorakis, E.E.; Doxaki, C.; Lagoudaki, E.; Ieronymaki, E.; Alexaki, V.I.; Helms, M.; Kondili, E.; Soennichsen, B.; et al. Akt2 deficiency protects from acute lung injury via alternative macrophage activation and miR-146a induction in mice. J. Immunol. 2014, 192, 394–406. [Google Scholar] [CrossRef]
- Wang, L.; Zhao, H.; Wang, L.; Tao, Y.; Du, G.; Guan, W.; Liu, J.; Brennan, C.; Ho, C.T.; Li, S. Effects of Selected Resveratrol Analogues on Activation and Polarization of Lipopolysaccharide-Stimulated BV-2 Microglial Cells. J. Agric. Food Chem. 2020, 68, 3750–3757. [Google Scholar] [CrossRef]
- Ge, Y.T.; Zhong, A.Q.; Xu, G.F.; Lu, Y. Resveratrol protects BV2 mouse microglial cells against LPS-induced inflammatory injury by altering the miR-146a-5p/TRAF6/NF-κB axis. Immun. Pharmacol. Immunotoxicol. 2019, 41, 549–557. [Google Scholar] [CrossRef]
- Juknat, A.; Gao, F.; Coppola, G.; Vogel, Z.; Kozela, E. miRNA expression profiles and molecular networks in resting and LPS-activated BV-2 microglia-Effect of cannabinoids. PLoS ONE 2019, 14, e0212039. [Google Scholar] [CrossRef] [PubMed]
- Gao, F.; Shen, J.; Zhao, L.; Hao, Q.; Yang, Y. Curcumin Alleviates Lipopolysaccharide (LPS)-Activated Neuroinflammation via Modulation of miR-199b-5p/IκB Kinase β (IKKβ)/Nuclear Factor Kappa B (NF-κB) Pathway in Microglia. Med. Sci. Monit. 2019, 25, 9801–9810. [Google Scholar] [CrossRef] [PubMed]
- Parisi, C.; Napoli, G.; Amadio, S.; Spalloni, A.; Apolloni, S.; Longone, P.; Volonté, C. MicroRNA-125b regulates microglia activation and motor neuron death in ALS. Cell Death Differ. 2016, 23, 531–541. [Google Scholar] [CrossRef] [Green Version]
- Aga, M.; Watters, J.J.; Pfeiffer, Z.A.; Wiepz, G.J.; Sommer, J.A.; Bertics, P.J. Evidence for nucleotide receptor modulation of cross talk between MAP kinase and NF-kappa B signaling pathways in murine RAW 264.7 macrophages. Am. J. Physiol. Cell Physiol. 2004, 286, C923–C930. [Google Scholar] [CrossRef]
- Ferrari, D.; Wesselborg, S.; Bauer, M.K.; Schulze-Osthoff, K. Extracellular ATP activates transcription factor NF-kappaB through the P2Z purinoreceptor by selectively targeting NF-kappaB p65. J. Cell Biol. 1997, 139, 1635–1643. [Google Scholar] [CrossRef]
- Parisi, C.; Arisi, I.; D’Ambrosi, N.; Storti, A.E.; Brandi, R.; D’Onofrio, M.; Volonté, C. Dysregulated microRNAs in amyotrophic lateral sclerosis microglia modulate genes linked to neuroinflammation. Cell Death Dis. 2013, 4, e959. [Google Scholar] [CrossRef]
- Veremeyko, T.; Yung, A.W.Y.; Dukhinova, M.; Strekalova, T.; Ponomarev, E.D. The Role of Neuronal Factors in the Epigenetic Reprogramming of Microglia in the Normal and Diseased Central Nervous System. Front. Cell Neurosci. 2019, 13, 453. [Google Scholar] [CrossRef] [Green Version]
- Ayata, P.; Badimon, A.; Strasburger, H.J.; Duff, M.K.; Montgomery, S.E.; Loh, Y.E.; Ebert, A.; Pimenova, A.A.; Ramirez, B.R.; Chan, A.T.; et al. Epigenetic regulation of brain region-specific microglia clearance activity. Nat. Neurosci. 2018, 21, 1049–1060. [Google Scholar] [CrossRef]
- Margueron, R.; Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 2011, 469, 343–349. [Google Scholar] [CrossRef] [Green Version]
- Swigut, T.; Wysocka, J. H3K27 Demethylases, at Long Last. Cell 2007, 131, 29–32. [Google Scholar] [CrossRef] [Green Version]
- Ponomarev, E.D.; Maresz, K.; Tan, Y.; Dittel, B.N. CNS-derived interleukin-4 is essential for the regulation of autoimmune inflammation and induces a state of alternative activation in microglial cells. J. Neurosci. 2007, 27, 10714–10721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Q.; Barres, B.A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 2018, 18, 225–242. [Google Scholar] [CrossRef] [PubMed]
- Elmore, M.R.P.; Najafi, A.R.; Koike, M.A.; Dagher, N.N.; Spangenberg, E.E.; Rice, R.A.; Kitazawa, M.; Matusow, B.; Nguyen, H.; West, B.L.; et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 2014, 82, 380–397. [Google Scholar] [CrossRef] [Green Version]
- Fourgeaud, L.; Través, P.G.; Tufail, Y.; Leal-Bailey, H.; Lew, E.D.; Burrola, P.G.; Callaway, P.; Zagórska, A.; Rothlin, C.V.; Nimmerjahn, A.; et al. TAM receptors regulate multiple features of microglial physiology. Nature 2016, 532, 240–244. [Google Scholar] [CrossRef] [Green Version]
- Goldmann, T.; Wieghofer, P.; Jordão, M.J.C.; Prutek, F.; Hagemeyer, N.; Frenzel, K.; Amann, L.; Staszewski, O.; Kierdorf, K.; Krueger, M.; et al. Origin, fate and dynamics of macrophages at central nervous system interfaces. Nat. Immunol. 2016, 17, 797–805. [Google Scholar] [CrossRef]
- Rothhammer, V.; Borucki, D.M.; Tjon, E.C.; Takenaka, M.C.; Chao, C.C.; Ardura-Fabregat, A.; de Lima, K.A.; Gutiérrez-Vázquez, C.; Hewson, P.; Staszewski, O.; et al. Microglial control of astrocytes in response to microbial metabolites. Nature 2018, 557, 724–728. [Google Scholar] [CrossRef]
- Gosselin, D.; Link, V.M.; Romanoski, C.E.; Fonseca, G.J.; Eichenfield, D.Z.; Spann, N.J.; Stender, J.D.; Chun, H.B.; Garner, H.; Geissmann, F.; et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 2014, 159, 1327–1340. [Google Scholar] [CrossRef]
- Holtman, I.R.; Skola, D.; Glass, C.K. Transcriptional control of microglia phenotypes in health and disease. J. Clin. Investig. 2017, 127, 3220–3229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheray, M.; Joseph, B. Epigenetics Control Microglia Plasticity. Front. Cell Neurosci. 2018, 12, 243. [Google Scholar] [CrossRef] [Green Version]
- de Groot, A.E.; Pienta, K.J. Epigenetic control of macrophage polarization: Implications for targeting tumor-associated macrophages. Oncotarget 2018, 9, 20908–20927. [Google Scholar] [CrossRef] [Green Version]
- Zhao, L.; Zabel, M.K.; Wang, X.; Ma, W.; Shah, P.; Fariss, R.N.; Qian, H.; Parkhurst, C.N.; Gan, W.B.; Wong, W.T. Microglial phagocytosis of living photoreceptors contributes to inherited retinal degeneration. EMBO Mol. Med. 2015, 7, 1179–1197. [Google Scholar] [CrossRef] [PubMed]
- Tikhanovich, I.; Zhao, J.; Olson, J.; Adams, A.; Taylor, R.; Bridges, B.; Marshall, L.; Roberts, B.; Weinman, S.A. Protein arginine methyltransferase 1 modulates innate immune responses through regulation of peroxisome proliferator-activated receptor γ-dependent macrophage differentiation. J. Biol. Chem. 2017, 292, 6882–6894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kittan, N.A.; Allen, R.M.; Dhaliwal, A.; Cavassani, K.A.; Schaller, M.; Gallagher, K.A.; Carson, W.F.; Mukherjee, S.; Grembecka, J.; Cierpicki, T.; et al. Cytokine induced phenotypic and epigenetic signatures are key to establishing specific macrophage phenotypes. PLoS ONE 2013, 8, e78045. [Google Scholar] [CrossRef] [Green Version]
- Tang, Y.; Li, T.; Li, J.; Yang, J.; Liu, H.; Zhang, X.J.; Le, W. Jmjd3 is essential for the epigenetic modulation of microglia phenotypes in the immune pathogenesis of Parkinson’s disease. Cell Death Differ. 2014, 21, 369–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carrier, M.; Robert, M.È.; Ibáñez, F.G.; Desjardins, M.; Tremblay, M.È. Imaging the Neuroimmune Dynamics Across Space and Time. Front. Neurosci. 2020, 14, 903. [Google Scholar] [CrossRef]
- Gabriel, E.M.; Fisher, D.T.; Evans, S.; Takabe, K.; Skitzki, J.J. Intravital microscopy in the study of the tumor microenvironment: From bench to human application. Oncotarget 2018, 9, 20165–20178. [Google Scholar] [CrossRef] [Green Version]
- Misgeld, T.; Kerschensteiner, M. In vivo imaging of the diseased nervous system. Nat. Rev. Neurosci. 2006, 7, 449–463. [Google Scholar] [CrossRef]
- Pittet, M.J.; Weissleder, R. Intravital imaging. Cell 2011, 147, 983–991. [Google Scholar] [CrossRef] [Green Version]
- Prunier, C.; Chen, N.; Ritsma, L.; Vrisekoop, N. Procedures and applications of long-term intravital microscopy. Methods 2017, 128, 52–64. [Google Scholar] [CrossRef]
- Weigert, R.; Sramkova, M.; Parente, L.; Amornphimoltham, P.; Masedunskas, A. Intravital microscopy: A novel tool to study cell biology in living animals. Histochem. Cell Biol. 2010, 133, 481–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lana, D.; Ugolini, F.; Giovannini, M.G. An Overview on the Differential Interplay among Neurons–Astrocytes–Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front. Cell Neurosci. 2020, 14, 585833. [Google Scholar] [CrossRef] [PubMed]
- Mittal, K.; Eremenko, E.; Berner, O.; Elyahu, Y.; Strominger, I.; Apelblat, D.; Nemirovsky, A.; Spiegel, I.; Monsonego, A. CD4 T Cells Induce A Subset of MHCII-Expressing Microglia that Attenuates Alzheimer Pathology. Science 2019, 16, 298–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Otxoa-de-Amezaga, A.; Miró-Mur, F.; Pedragosa, J.; Gallizioli, M.; Justicia, C.; Gaja-Capdevila, N.; Ruíz-Jaen, F.; Salas-Perdomo, A.; Bosch, A.; Calvo, M.; et al. Microglial cell loss after ischemic stroke favors brain neutrophil accumulation. Acta Neuropathol. 2019, 137, 321–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Graf, B.W.; Boppart, S.A. Imaging and analysis of three-dimensional cell culture models. Methods Mol. Biol. 2010, 591, 211–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hierro-Bujalance, C.; Bacskai, B.J.; Garcia-Alloza, M. In vivo imaging of microglia with multiphoton microscopy. Front. Aging Neurosci. 2018, 10, 218. [Google Scholar] [CrossRef]
- Neumann, J.; Henneberg, S.; Von Kenne, S.; Nolte, N.; Müller, A.J.; Schraven, B.; Görtler, M.W.; Reymann, K.G.; Gunzer, M.; Riek-Burchardt, M. Beware the intruder: Real time observation of infiltrated neutrophils and neutrophil-Microglia interaction during stroke in vivo. PLoS ONE 2018, 13, e0193970. [Google Scholar] [CrossRef] [Green Version]
- Savage, J.C.; Carrier, M.; Tremblay, M.È. Morphology of Microglia Across Contexts of Health and Disease. Methods Mol. Biol. 2019, 2034, 13–26. [Google Scholar] [CrossRef]
- Li, Y.; Du, X.F.; Liu, C.S.; Wen, Z.L.; Du, J.L. Reciprocal Regulation between Resting Microglial Dynamics and Neuronal Activity In vivo. Dev. Cell 2012, 23, 1189–1202. [Google Scholar] [CrossRef] [Green Version]
- Wake, H.; Moorhouse, A.J.; Jinno, S.; Kohsaka, S.; Nabekura, J. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J. Neurosci. 2009, 29, 3974–3980. [Google Scholar] [CrossRef] [Green Version]
- Chen, Z.; Ross, J.L.; Hambardzumyan, D. Intravital 2-photon imaging reveals distinct morphology and infiltrative properties of glioblastoma-associated macrophages. Proc. Natl. Acad. Sci. USA 2019, 116, 14254–14259. [Google Scholar] [CrossRef] [Green Version]
- Bethge, P.; Chéreau, R.; Avignone, E.; Marsicano, G.; Nägerl, U.V. Two-photon excitation STED microscopy in two colors in acute brain slices. Biophys. J. 2013, 104, 778–785. [Google Scholar] [CrossRef] [Green Version]
- Evans, T.A.; Barkauskas, D.S.; Myers, J.T.; Huang, A.Y. Intravital imaging of axonal interactions with microglia and macrophages in a mouse dorsal column crush injury. J. Vis. Exp. 2014, 23, e52228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fekete, R.; Cserép, C.; Lénárt, N.; Tóth, K.; Orsolits, B.; Martinecz, B.; Méhes, E.; Szabó, B.; Németh, V.; Gönci, B.; et al. Microglia control the spread of neurotropic virus infection via P2Y12 signalling and recruit monocytes through P2Y12-independent mechanisms. Acta Neuropathol. 2018, 136, 461–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qiao, S.; Qian, Y.; Xu, G.; Luo, Q.; Zhang, Z. Long-term characterization of activated microglia/macrophages facilitating the development of experimental brain metastasis through intravital microscopic imaging. J. Neuroinflamm. 2019, 16, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tremblay, M.Ě.; Lowery, R.L.; Majewska, A.K. Microglial interactions with synapses are modulated by visual experience. PLoS Biol. 2010, 8, e1000527. [Google Scholar] [CrossRef] [Green Version]
- Heintzmann, R.; Huser, T. Super-Resolution Structured Illumination Microscopy. Chem. Rev. 2017, 117, 13890–13908. [Google Scholar] [CrossRef]
- Hahn, C.; Becker, K.; Saghafi, S.; Pende, M.; Avdibašić, A.; Foroughipour, M.; Heinz, D.E.; Wotjak, C.T.; Dodt, H.U. High-resolution imaging of fluorescent whole mouse brains using stabilised organic media (sDISCO). J. Biophotonics 2019, 12, e368. [Google Scholar] [CrossRef] [Green Version]
- Qi, Y.; Yu, T.; Xu, J.; Wan, P.; Ma, Y.; Zhu, J.; Li, Y.; Gong, H.; Luo, Q.; Zhu, D. FDISCO: Advanced solvent-based clearing method for imaging whole organs. Sci. Adv. 2019, 5, e8355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ormel, P.R.; de Sá, R.V.; van Bodegraven, E.J.; Karst, H.; Harschnitz, O.; Sneeboer, M.A.M.; Johansen, L.E.; van Dijk, R.E.; Scheefhals, N.; van Berlekom, A.B.; et al. Microglia innately develop within cerebral organoids. Nat. Commun. 2018, 9, 4167. [Google Scholar] [CrossRef]
- Pfeiffer, T.; Poll, S.; Bancelin, S.; Angibaud, J.; Inavalli, V.V.G.K.; Keppler, K.; Mittag, M.; Fuhrmann, M.; Nägerl, U.V. Chronic 2P-STED imaging reveals high turnover of dendritic spines in the hippocampus in vivo. eLife 2018, 7, e34700. [Google Scholar] [CrossRef]
- Vangindertael, J.; Camacho, R.; Sempels, W.; Mizuno, H.; Dedecker, P.; Janssen, K.P.F. An introduction to optical super-resolution microscopy for the adventurous biologist. Methods Appl. Fluoresc. 2018, 6, 022003. [Google Scholar] [CrossRef] [PubMed]
- Cserép, C.; Pósfai, B.; Lénárt, N.; Fekete, R.; László, Z.I.; Lele, Z.; Orsolits, B.; Molnár, G.; Heindl, S.; Schwarcz, A.D.; et al. Microglia monitor and protect neuronal function via specialized somatic purinergic junctions. Science 2020, 367, 528–537. [Google Scholar] [CrossRef]
- Varga, D.P.; Menyhárt, Á.; Pósfai, B.; Császár, E.; Lénárt, N.; Cserép, C.; Orsolits, B.; Martinecz, B.; Szlepák, T.; Bari, F.; et al. Microglia alter the threshold of spreading depolarization and related potassium uptake in the mouse brain. J. Cereb Blood Flow Metab. 2020, 40, S67–S80. [Google Scholar] [CrossRef]
- Fumagalli, S.; Fiordaliso, F.; Perego, C.; Corbelli, A.; Mariani, A.; De Paola, M.; De Simoni, M.G. The phagocytic state of brain myeloid cells after ischemia revealed by superresolution structured illumination microscopy. J. Neuroinflamm. 2019, 16, 9. [Google Scholar] [CrossRef] [Green Version]
- Schafer, D.P.; Lehrman, E.K.; Kautzman, A.G.; Koyama, R.; Mardinly, A.R.; Yamasaki, R.; Ransohoff, R.M.; Greenberg, M.E.; Barres, B.A.; Stevens, B. Microglia Sculpt Postnatal Neural Circuits in an Activity and Complement-Dependent Manner. Neuron 2012, 74, 691–705. [Google Scholar] [CrossRef] [Green Version]
- Beckman, D.; Ott, S.; Donis-Cox, K.; Janssen, W.G.; Bliss-Moreau, E.; Rudebeck, P.H.; Baxter, M.G.; Morrison, J.H. Oligomeric Aβ in the monkey brain impacts synaptic integrity and induces accelerated cortical aging. Proc. Natl. Acad. Sci. USA 2019, 116, 26239–26246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Konishi, H.; Okamoto, T.; Hara, Y.; Komine, O.; Tamada, H.; Maeda, M.; Osako, F.; Kobayashi, M.; Nishiyama, A.; Kataoka, Y.; et al. Astrocytic phagocytosis is a compensatory mechanism for microglial dysfunction. EMBO J. 2020, 39, e104464. [Google Scholar] [CrossRef]
- Weinhard, L.; Di Bartolomei, G.; Bolasco, G.; Machado, P.; Schieber, N.L.; Neniskyte, U.; Exiga, M.; Vadisiute, A.; Raggioli, A.; Schertel, A.; et al. Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction. Nat. Commun. 2018, 9, 1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karreman, M.A.; Hyenne, V.; Schwab, Y.; Goetz, J.G. Intravital Correlative Microscopy: Imaging Life at the Nanoscale. Trends Cell Biol. 2016, 26, 848–863. [Google Scholar] [CrossRef]
- Van Ham, T.J.; Brady, C.A.; Kalicharan, R.D.; Oosterhof, N.; Kuipers, J.; Veenstra-Algra, A.; Sjollema, K.A.; Peterson, R.T.; Kampinga, H.H.; Giepmans, B.N.G. Intravital correlated microscopy reveals differential macrophage and microglial dynamics during resolution of neuroinflammation. Dis. Model Mech. 2014, 7, 857–869. [Google Scholar] [CrossRef] [Green Version]
- Luckner, M.; Burgold, S.; Filser, S.; Scheungrab, M.; Niyaz, Y.; Hummel, E.; Wanner, G.; Herms, J. Label-free 3D-CLEM Using Endogenous Tissue Landmarks. Science 2018, 6, 92–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cunningham, C.L.; Martínez-Cerdeño, V.; Noctor, S.C. Microglia Regulate the Number of Neural Precursor Cells in the Developing Cerebral Cortex. J. Neurosci. 2013, 33, 4216–4233. [Google Scholar] [CrossRef] [Green Version]
- Miyamoto, A.; Wake, H.; Moorhouse, A.J.; Nabekura, J. Microglia and Synapse Interactions: Fine Tuning Neural Circuits and Candidate Molecules. Front. Cell Neurosci. 2013, 7, 70. [Google Scholar] [CrossRef] [Green Version]
- Kettenmann, H.; Kirchhoff, F.; Verkhratsky, A. Microglia: New Roles for the Synaptic Stripper. Neuron 2013, 77, 10–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Watson, P.M.D.; Kavanagh, E.; Allenby, G.; Vassey, M. Bioengineered 3D Glial Cell Culture Systems and Applications for Neurodegeneration and Neuroinflammation. SLAS Discov. 2017, 22, 583–601. [Google Scholar] [CrossRef] [Green Version]
- Pöttler, M.; Zierler, S.; Kerschbaum, H.H. An Artificial Three-Dimensional Matrix Promotes Ramification in the Microglial Cell-Line, BV-2. Neurosci. Lett. 2006, 410, 137–140. [Google Scholar] [CrossRef]
- Cho, H.J.; Verbridge, S.S.; Davalos, R.V.; Lee, Y.W. Development of an In vitro 3D Brain Tissue Model Mimicking In vivo-Like Pro-Inflammatory and Pro-Oxidative Responses. Ann. Biomed. Eng. 2018, 46, 877–887. [Google Scholar] [CrossRef]
- Haw, R.T.Y.; Tong, C.K.; Yew, A.; Lee, H.C.; Phillips, J.B.; Vidyadaran, S. A Three-Dimensional Collagen Construct to Model Lipopolysaccharide-Induced Activation of BV2 Microglia. J. Neuroinflamm. 2014, 11, 134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chronopoulou, L.; Togna, A.R.; Guarguaglini, G.; Masci, G.; Giammaruco, F.; Togna, G.I.; Palocci, C. Self-Assembling Peptide Hydrogels Promote Microglial Cells Proliferation and NGF Production. Soft Matter 2012, 8, 5784–5790. [Google Scholar] [CrossRef]
- Park, J.; Wetzel, I.; Marriott, I.; Dréau, D.; D’Avanzo, C.; Kim, D.Y.; Tanzi, R.E.; Cho, H. A 3D Human Tri-Culture System Modeling Neurodegeneration and Neuroinflammation in Alzheimer’s Disease. Nat. Neurosci. 2018, 21, 941–951. [Google Scholar] [CrossRef]
- Cai, H.; Ao, Z.; Hu, L.; Moon, Y.; Wu, Z.; Lu, H.C.; Kim, J.; Guo, F. Acoustofluidic Assembly of 3D Neurospheroids to Model Alzheimer’s Disease. Analyst 2020, 145, 6243–6253. [Google Scholar] [CrossRef]
- Lancaster, M.A.; Knoblich, J.A. Organogenesis in a Dish: Modeling Development and Disease Using Organoid Technologies. Science 2014, 345, 1247125. [Google Scholar] [CrossRef] [PubMed]
- Lancaster, M.A.; Knoblich, J.A. Generation of Cerebral Organoids from Human Pluripotent Stem Cells. Nat. Protoc. 2014, 9, 2329–2340. [Google Scholar] [CrossRef] [Green Version]
- Dixit, C.K.; Kaushik, A. Microfluidics for Biologists: Fundamentals and Applications; Springer International Publishing Switzerland: Cham, Switzerland, 2016; ISBN 978-3-319-40036-5. [Google Scholar] [CrossRef]
- Park, J.W.; Vahidi, B.; Taylor, A.M.; Rhee, S.W.; Jeon, N.L. Microfluidic Culture Platform for Neuroscience Research. Nat. Protoc. 2006, 1, 2128–2136. [Google Scholar] [CrossRef] [PubMed]
- Gross, P.G.; Kartalov, E.P.; Scherer, A.; Weiner, L.P. Applications of Microfluidics for Neuronal Studies. J. Neurol. Sci. 2007, 252, 135–143. [Google Scholar] [CrossRef] [PubMed]
- Campenot, R.B. Local Control of Neurite Development by Nerve Growth Factor. Proc. Natl. Acad. Sci. USA 1977, 74, 4516–4519. [Google Scholar] [CrossRef] [Green Version]
- Raff, M.C.; Whitmore, A.V.; Finn, J.T. Axonal self-destruction and neurodegeneration. Science 2002, 296, 868–871. [Google Scholar] [CrossRef] [PubMed]
- Zweifel, L.S.; Kuruvilla, R.; Ginty, D.D. Functions and Mechanisms of Retrograde Neurotrophin Signalling. Nat. Rev. Neurosci. 2005, 6, 615–625. [Google Scholar] [CrossRef]
- Taylor, A.M.; Rhee, S.W.; Tu, C.H.; Cribbs, D.H.; Cotman, C.W.; Jeon, N.L. Microfluidic Multicompartment Device for Neuroscience Research. Langmuir 2003, 19, 1551–1556. [Google Scholar] [CrossRef] [Green Version]
- Park, J.; Koito, H.; Li, J.; Han, A. A Multi-Compartment CNS Neuron-Glia Co-Culture Microfluidic Platform. J. Vis. Exp. 2009, 10, 1399. [Google Scholar] [CrossRef]
- Rhee, S.W.; Taylor, A.M.; Tu, C.H.; Cribbs, D.H.; Cotman, C.W.; Jeon, N.L. Patterned Cell Culture inside Microfluidic Devices. Lab. Chip. 2005, 5, 102–107. [Google Scholar] [CrossRef] [PubMed]
- Taylor, A.M.; Blurton-Jones, M.; Rhee, S.W.; Cribbs, D.H.; Cotman, C.W.; Jeon, N.L. A Microfluidic Culture Platform for CNS Axonal Injury, Regeneration and Transport. Nat. Methods. 2005, 2, 599–605. [Google Scholar] [CrossRef] [PubMed]
- Hosmane, S.; Yang, I.H.; Ruffin, A.; Thakor, N.; Venkatesan, A. Circular Compartmentalized Microfluidic Platform: Study of Axon-Glia Interactions. Lab Chip 2010, 10, 741–747. [Google Scholar] [CrossRef]
- Hosmane, S.; Tegenge, M.A.; Rajbhandari, L.; Uapinyoying, P.; Kumar, N.G.; Thakor, N.; Venkatesan, A. Toll/Interleukin-1 Receptor Domain-Containing Adapter Inducing Interferon-β Mediates Microglial Phagocytosis of Degenerating Axons. J. Neurosci. 2012, 32, 7745–7757. [Google Scholar] [CrossRef] [Green Version]
- Majumdar, D.; Gao, Y.; Li, D.; Webb, D.J. Co-Culture of Neurons and Glia in a Novel Microfluidic Platform. J. Neurosci. Methods 2011, 196, 38–44. [Google Scholar] [CrossRef] [Green Version]
- Shi, M.; Majumdar, D.; Gao, Y.; Brewer, B.M.; Goodwin, C.R.; McLean, J.A.; Li, D.; Webb, D.J. Glia Co-Culture with Neurons in Microfluidic Platforms Promotes the Formation and Stabilization of Synaptic Contacts. Lab. Chip. 2013, 13, 3008–3021. [Google Scholar] [CrossRef] [Green Version]
- Fujita, Y.; Nakanishi, T.; Ueno, M.; Itohara, S.; Yamashita, T. Netrin-G1 Regulates Microglial Accumulation along Axons and Supports the Survival of Layer V Neurons in the Postnatal Mouse Brain. Cell Rep. 2020, 31, 107580. [Google Scholar] [CrossRef]
- Fujita, Y.; Yamashita, T. Protocol for Co-Culture of Microglia with Axons. STAR Protoc. 2020, 15, 100111. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Petralla, S.; De Chirico, F.; Miti, A.; Tartagni, O.; Massenzio, F.; Poeta, E.; Virgili, M.; Zuccheri, G.; Monti, B. Epigenetics and Communication Mechanisms in Microglia Activation with a View on Technological Approaches. Biomolecules 2021, 11, 306. https://doi.org/10.3390/biom11020306
Petralla S, De Chirico F, Miti A, Tartagni O, Massenzio F, Poeta E, Virgili M, Zuccheri G, Monti B. Epigenetics and Communication Mechanisms in Microglia Activation with a View on Technological Approaches. Biomolecules. 2021; 11(2):306. https://doi.org/10.3390/biom11020306
Chicago/Turabian StylePetralla, Sabrina, Francesca De Chirico, Andrea Miti, Ottavia Tartagni, Francesca Massenzio, Eleonora Poeta, Marco Virgili, Giampaolo Zuccheri, and Barbara Monti. 2021. "Epigenetics and Communication Mechanisms in Microglia Activation with a View on Technological Approaches" Biomolecules 11, no. 2: 306. https://doi.org/10.3390/biom11020306
APA StylePetralla, S., De Chirico, F., Miti, A., Tartagni, O., Massenzio, F., Poeta, E., Virgili, M., Zuccheri, G., & Monti, B. (2021). Epigenetics and Communication Mechanisms in Microglia Activation with a View on Technological Approaches. Biomolecules, 11(2), 306. https://doi.org/10.3390/biom11020306