Macrophage–Neuroglia Interactions in Promoting Neuronal Regeneration in Zebrafish
Abstract
:1. Introduction
2. Overview of the Response of Macrophages to Spinal Cord Injury in Zebrafish
2.1. Phase 1: Acute Inflammatory Response
2.2. Phase 2: Resolution of Inflammation
2.3. Phase 3: Promotion of Axon Regeneration
2.4. Phase 4: Modulation of Glial Scar Formation
3. The Role of TNF-Activated Macrophages in Pro-Regenerative Neurogenesis
4. The TNF/Tnfrsf1a Mediated AP-1 Signaling Pathway Increases Hdac1 after Injury
5. Macrophages Promote Neuronal Regeneration through ERG Progenitor Cells after Injury
6. Macrophage TNF-α Signaling May Regulate pMN Progenitor Cell Fate
7. Macrophage TNF-α Signaling May Regulate Radial Glial Cell-Mediated Neurogenesis
8. Conclusions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Shechter, R.; Miller, O.; Yovel, G.; Rosenzweig, N.; London, A.; Ruckh, J.; Kim, K.; Klein, E.; Kalchenko, V.; Bendel, P.; et al. Recruitment of beneficial m2 macrophages to injured spinal cord is orchestrated by remote brain choroid plexus. Immunity 2013, 38, 555–569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gensel, J.; Nakamura, S.; Guan, Z.; van Rooijen, N.; Ankeny, D.; Popovich, P. Macrophages promote axon regeneration with concurrent neurotoxicity. J. Neurosci. 2009, 29, 3956–3968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nguyen-Chi, M.; Laplace-Builhe, B.; Travnickova, J.; Luz-Crawford, P.; Tejedor, G.; Phan, Q.; Duroux-Richard, I.; Levraud, J.; Kissa, K.; Lutfalla, G.; et al. Identification of polarized macrophage subsets in zebrafish. eLife 2015, 4, e07288. [Google Scholar] [CrossRef] [PubMed]
- Becker, T.; Becker, C.G. Regenerating descending axons preferentially reroute to the gray matter in the presence of a general macrophage/microglial reaction caudal to a spinal transection in adult zebrafish. J. Comp. Neurol. 2001, 433, 131–147. [Google Scholar] [CrossRef]
- Ohnmacht, J.; Yang, Y.; Maurer, G.W.; Barreiro-Iglesias, A.; Tsarouchas, T.M.; Wehner, D.; Sieger, D.; Becker, C.G.; Becker, T. Spinal motor neurons are regenerated after mechanical lesion and genetic ablation in larval zebrafish. Development 2016, 143, 1464–1474. [Google Scholar] [CrossRef] [Green Version]
- Hui, E.; Cheung, J.; Zhu, J.; Su, X.; Taylor, M.J.; Wallweber, H.A.; Sasmal, D.K.; Huang, J.; Kim, J.M.; Mellman, I.; et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017, 355, 1428–1433. [Google Scholar] [CrossRef]
- Al-Gayyar, M.M.; Elsherbiny, N.M. Contribution of TNF-α to the development of retinal neurodegenerative disorders. Eur. Cytokine Netw. 2013, 24, 27–36. [Google Scholar] [CrossRef] [Green Version]
- Wan, J.; Ramachandran, R.; Goldman, D. HB-EGF is necessary and sufficient for Müller glia dedifferentiation and retina regeneration. Dev. Cell 2012, 22, 334–347. [Google Scholar] [CrossRef] [Green Version]
- Beldi, G.; Khosravi, M.; Abdelgawad, M.E.; Salomon, B.L.; Uzan, G.; Haouas, H.; Naserian, S. TNFα/TNFR2 signaling pathway: An active immune checkpoint for mesenchymal stem cell immunoregulatory function. Stem Cell Res. Ther. 2020, 11, 281. [Google Scholar] [CrossRef]
- Tsarouchas, T.; Wehner, D.; Cavone, L.; Munir, T.; Keatinge, M.; Lambertus, M.; Underhill, A.; Barrett, T.; Kassapis, E.; Ogryzko, N.; et al. Dynamic control of proinflammatory cytokines Il-1β and Tnf-α by macrophages in zebrafish spinal cord regeneration. Nat. Commun. 2018, 9, 4670. [Google Scholar] [CrossRef] [Green Version]
- Cavone, L.; McCann, T.; Drake, L.K.; Aguzzi, E.A.; Oprişoreanu, A.M.; Pedersen, E.; Sandi, S.; Selvarajah, J.; Tsarouchas, T.M.; Wehner, D.; et al. A unique macrophage subpopulation signals directly to progenitor cells to promote regenerative neurogenesis in the zebrafish spinal cord. Dev. Cell 2021, 56, 1617–1630. [Google Scholar] [CrossRef] [PubMed]
- Briona, L.K.; Poulain, F.E.; Mosimann, C.; Dorsky, R.I. Wnt/ß-catenin signaling is required for radial glial neurogenesis following spinal cord injury. Dev. Biol. 2015, 403, 15–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goldshmit, Y.; Tang, J.K.K.; Siegel, A.L.; Nguyen, P.D.; Kaslin, J.; Currie, P.D.; Jusuf, P.R. Different Fgfs have distinct roles in regulating neurogenesis after spinal cord injury in zebrafish. Neural Dev. 2018, 13, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, L.; Fu, C.; Xiong, F.; He, C.; Wei, Q. Stem cell therapy for spinal cord injury. Cell Transplant. 2021, 30, 963689721989266. [Google Scholar] [CrossRef] [PubMed]
- Zeng, C.W.; Kamei, Y.; Shigenobu, S.; Sheu, J.C.; Tsai, H.J. Injury-induced Cavl-expressing cells at lesion rostral side play major roles in spinal cord regeneration. Open Biol. 2021, 11, 200304. [Google Scholar] [CrossRef]
- Var, S.R.; Byrd-Jacobs, C.A. Role of macrophages and microglia in zebrafish regeneration. Int. J. Mol. Sci. 2020, 21, 4768. [Google Scholar] [CrossRef]
- Kanazawa, M.; Ninomiya, I.; Hatakeyama, M.; Takahashi, T.; Shimohata, T. Microglia and monocytes/macrophages polarization reveal novel therapeutic mechanism against stroke. Int. J. Mol. Sci. 2017, 18, 2135. [Google Scholar] [CrossRef] [Green Version]
- Xiong, X.Y.; Liu, L.; Yang, Q.W. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog. Neurobiol. 2016, 142, 23–44. [Google Scholar] [CrossRef]
- Ma, P.F.; Gao, C.C.; Yi, J.; Zhao, J.L.; Liang, S.Q.; Zhao, Y.; Ye, Y.-C.; Bai, J.; Zheng, Q.-J.; Dou, K.-F.; et al. Cytotherapy with M1-polarized macrophages ameliorates liver fibrosis by modulating immune microenvironment in mice. J. Hepatol. 2017, 67, 770–779. [Google Scholar] [CrossRef]
- Sipe, G.O.; Lowery, R.L.; Tremblay, M.È.; Kelly, E.A.; Lamantia, C.E.; Majewska, A.K. Microglial P2Y12 is necessary for synaptic plasticity in mouse visual cortex. Nat. Commun. 2016, 7, 10905. [Google Scholar] [CrossRef]
- Lee, C.H.; Chun, T. Anti-inflammatory role of TAM family of receptor tyrosine kinases via modulating macrophage function. Mol. Cells 2019, 42, 1. [Google Scholar] [PubMed]
- Chang, F.; Wang, Y.; Liu, P.; Peng, J.; Han, G.H.; Ding, X.; Wei, S.; Gao, G.; Huang, K. Role of macrophages in peripheral nerve injury and repair. Neural Regen. Res. 2019, 14, 1335. [Google Scholar] [CrossRef] [PubMed]
- Yu, Y.; Yue, Z.; Xu, M.; Zhang, M.; Shen, X.; Ma, Z.; Li, J.; Xie, X. Macrophages play a key role in tissue repair and regeneration. PeerJ 2022, 10, e14053. [Google Scholar] [CrossRef] [PubMed]
- Petrie, T.A.; Strand, N.S.; Tsung-Yang, C.; Rabinowitz, J.S.; Moon, R.T. Macrophages modulate adult zebrafish tail fin regeneration. Development 2014, 141, 2581–2591. [Google Scholar] [CrossRef] [PubMed]
- Majidinia, M.; Aghazadeh, J.; Jahanban-Esfahlani, R.; Yousefi, B. The roles of Wnt/β-catenin pathway in tissue development and regenerative medicine. J. Cell. Physiol. 2018, 2338, 5598–5612. [Google Scholar] [CrossRef] [PubMed]
- Lucini, C.; D’Angelo, L.; Cacialli, P.; Palladino, A.; De Girolamo, P. BDNF, brain, and regeneration: Insights from zebrafish. Int. J. Mol. Sci. 2018, 19, 3155. [Google Scholar] [CrossRef] [Green Version]
- Tayanloo-Beik, A.; Rabbani, Z.; Soveyzi, F.; Alavi-Moghadam, S.; Rezaei-Tavirani, M.; Goodarzi, P.; Arjmand, B.; Larijani, B. Cellular therapy for treatment of spinal cord injury in Zebrafish model. Mol. Biol. Rep. 2021, 48, 1787–1800. [Google Scholar] [CrossRef]
- Li, R.; Li, D.H.; Zhang, H.Y.; Wang, J.; Li, X.K.; Xiao, J. Growth factors-based therapeutic strategies and their underlying signaling mechanisms for peripheral nerve regeneration. Acta Pharmacol. Sin. 2020, 41, 1289–1300. [Google Scholar] [CrossRef]
- Tarnawski, A.S.; Ahluwalia, A. The critical role of growth factors in gastric ulcer healing: The cellular and molecular mechanisms and potential clinical implications. Cells 2021, 10, 1964. [Google Scholar] [CrossRef]
- Adams, K.L.; Gallo, V. The diversity and disparity of the glial scar. Nat. Neurosci. 2018, 21, 9–15. [Google Scholar] [CrossRef]
- Aurora, A.B.; Olson, E.N. Immune modulation of stem cells and regeneration. Cell Stem Cell 2014, 15, 14–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silver, J.; Miller, J.H. Regeneration beyond the glial scar. Nat. Rev. Neurosci. 2004, 5, 146–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liddelow, S.A.; Barres, B.A. Not everything is scary about a glial scar. Nature 2016, 532, 182–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sabin, K.Z.; Echeverri, K. The role of the immune system during regeneration of the central nervous system. J. Immunol. Regen. Med. 2020, 7, 100023. [Google Scholar] [CrossRef]
- Lyu, J.; Xie, D.; Bhatia, T.N.; Leak, R.K.; Hu, X.; Jiang, X. Microglial/Macrophage polarization and function in brain injury and repair after stroke. CNS Neurosci. Ther. 2021, 27, 515–527. [Google Scholar] [CrossRef]
- Hunyara, J.L.; Kolodkin, A.L. Repurposing developmental mechanisms in the adult nervous system. Curr. Opin. Genet. Dev. 2020, 65, 14–21. [Google Scholar] [CrossRef]
- Cruceriu, D.; Baldasici, O.; Balacescu, O.; Berindan-Neagoe, I. The dual role of tumor necrosis factor-alpha (TNF-α) in breast cancer: Molecular insights and therapeutic approaches. Cell. Oncol. 2020, 43, 1–18. [Google Scholar] [CrossRef]
- Bosak, V.; Murata, K.; Bludau, O.; Brand, M. Role of the immune response in initiating central nervous system regeneration in vertebrates: Learning from the fish. Int. J. Dev. Biol. 2018, 62, 403–417. [Google Scholar] [CrossRef] [Green Version]
- Yong, H.Y.; Rawji, K.S.; Ghorbani, S.; Xue, M.; Yong, V.W. The benefits of neuroinflammation for the repair of the injured central nervous system. Cell. Mol. Immunol. 2019, 16, 540–546. [Google Scholar] [CrossRef]
- Wen, X.; Jiao, L.; Tan, H. MAPK/ERK pathway as a central regulator in vertebrate organ regeneration. Int. J. Mol. Sci. 2022, 23, 1464. [Google Scholar] [CrossRef]
- Jin, M.; Opalek, J.M.; Marsh, C.B.; Wu, H.M. Proteome comparison of alveolar macrophages with monocytes reveals distinct protein characteristics. Am. J. Respir. Cell Mol. Biol. 2004, 31, 322–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amiel, A.R.; Tsai, S.L.; Wehner, D. Embracing the diversity of model systems to deconstruct the basis of regeneration and tissue repair. Development 2023, 150, dev201579. [Google Scholar] [CrossRef] [PubMed]
- Bernardino, L.; Agasse, F.; Silva, B.; Ferreira, R.; Grade, S.; Malva, J.O. Tumor necrosis factor-α modulates survival, proliferation, and neuronal differentiation in neonatal subventricular zone cell cultures. Stem Cells 2008, 26, 2361–2371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, L.; Xing, Q.; Huang, T.; Zhou, J.; Liu, T.; Cui, Y.; Cheng, T.; Wang, Y.; Zhou, X.; Yang, B.; et al. derived mesenchymal stem cells in a mouse model of traumatic brain injury via PI3K/AKT pathway. Front. Cell. Neurosci. 2019, 12, 498. [Google Scholar] [CrossRef] [Green Version]
- Belenguer, G.; Duart-Abadia, P.; Jordán-Pla, A.; Domingo-Muelas, A.; Blasco-Chamarro, L.; Ferrón, S.R.; Morante-Redolat, J.M.; Fariñas, I. Adult neural stem cells are alerted by systemic inflammation through TNF-α receptor signaling. Cell Stem Cell 2021, 28, 285–299. [Google Scholar] [CrossRef]
- Nguyen-Chi, M.; Laplace-Builhé, B.; Travnickova, J.; Luz-Crawford, P.; Tejedor, G.; Lutfalla, G.; Kissa, K.; Jorgensen, C.; Djouad, F. TNF signaling and macrophages govern fin regeneration in zebrafish larvae. Cell Death Dis. 2017, 8, e2979. [Google Scholar] [CrossRef] [Green Version]
- Lei, X.D.; Sun, Y.; Cai, S.J.; Fang, Y.W.; Cui, J.L.; Li, Y.H. Role of tumor necrosis factor-alpha in zebrafish retinal neurogenesis and myelination. Int. J. Ophthalmol. 2016, 9, 831–837. [Google Scholar]
- Himmler, A.; Maurer-Fogy, I.; Krönke, M.; Scheurich, P.; Pfizenmaier, K.; Lantz, M.; Olsson, I.; Hauptmann, R.; Stratowa, C.; Adolf, G. Molecular cloning and expression of human and rat tumor necrosis factor receptor chain (p60) and its soluble derivative, tumor necrosis factor-binding protein. DNA Cell Biol. 1990, 9, 705–715. [Google Scholar] [CrossRef]
- Sakurai, H.; Sugita, T. C-Jun N-terminal kinase-mediated AP-1 activation in experimental glomerulonephritis in rats. IUBMB Life 1998, 45, 831–839. [Google Scholar] [CrossRef]
- Choi, H.; Dikalova, A.; Stark, R.J.; Lamb, F.S. c-Jun N-terminal kinase attenuates TNFα signaling by reducing Nox1-dependent endosomal ROS production in vascular smooth muscle cells. Free Radic. Biol. Med. 2015, 86, 219–227. [Google Scholar] [CrossRef]
- Raivich, G.; Bohatschek, M.; Da Costa, C.; Iwata, O.; Galiano, M.; Hristova, M.; Nateri, A.S.; Makwana, M.; Riera-Sans, L.; Wolfer, D.P.; et al. The AP-1 transcription factor c-Jun is required for efficient axonal regeneration. Neuron 2004, 43, 57–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Montgomery, R.; Hsieh, J.; Barbosa, A.; Richardson, J.; Olson, E. Histone deacetylases 1 and 2 control the progression of neural precursors to neurons during brain development. Proc. Natl. Acad. Sci. USA 2009, 106, 7876–7881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cunliffe, V.T. Histone deacetylase 1 is required to repress Notch target gene expression during zebrafish neurogenesis and to maintain the production of motoneurones in response to hedgehog signalling. Development 2004, 131, 2983–2995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamaguchi, M.; Tonou-Fujimori, N.; Komori, A.; Maeda, R.; Nojima, Y.; Li, H.; Okamoto, H.; Masai, I. Histone deacetylase 1 regulates retinal neurogenesis in zebrafish by suppressing Wnt and Notch signaling pathways. Development 2005, 132, 3027–3043. [Google Scholar] [CrossRef] [Green Version]
- Mitchell, P.; Liew, G.; Gopinath, B.; Wong, T.Y. Age-related macular degeneration. Lancet 2018, 392, 1147–1159. [Google Scholar] [CrossRef]
- Ravanelli, A.; Appel, B. Motor neurons and oligodendrocytes arise from distinct cell lineages by progenitor recruitment. Genes Dev. 2015, 29, 2504–2515. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Q.; Choi, G.; Anderson, D. The bHLH Transcription factor Olig2 promotes oligodendrocyte differentiation in collaboration with Nkx2.2. Neuron 2001, 31, 791–807. [Google Scholar] [CrossRef] [Green Version]
- Li, H.; de Faria, J.P.; Andrew, P.; Nitarska, J.; Richardson, W.D. Phosphorylation regulates OLIG2 cofactor choice and the motor neuron-oligodendrocyte fate switch. Neuron 2011, 69, 918–929. [Google Scholar] [CrossRef] [Green Version]
- Dawson, M. NG2-expressing glial progenitor cells: An abundant and widespread population of cycling cells in the adult rat CNS. Mol. Cell. Neurosci. 2003, 24, 476–488. [Google Scholar] [CrossRef]
- Lee, S.K.; Lee, B.; Ruiz, E.C.; Pfaff, S.L. Olig2 and Ngn2 function in opposition to modulate gene expression in motor neuron progenitor cells. Genes Dev. 2005, 19, 282–294. [Google Scholar] [CrossRef] [Green Version]
- Scott, K.; O’Rourke, R.; Gillen, A.; Appel, B. Prdm8 regulates pMN progenitor specification for motor neuron and oligodendrocyte fates by modulating the Shh signaling response. Development 2020, 147, dev191023. [Google Scholar] [CrossRef] [PubMed]
- Doll, C.A.; Scott, K.; Appel, B. Fmrp regulates oligodendrocyte lineage cell specification and differentiation. Glia 2021, 69, 2349–2361. [Google Scholar] [CrossRef] [PubMed]
- Rabadán, M.A.; Cayuso, J.; Le Dréau, G.; Cruz, C.; Barzi, M.; Pons, S.; Briscoe, J.; Martí, E. Jagged 2 controls the generation of motor neuron and oligodendrocyte progenitors in the ventral spinal cord. Cell Death Differ. 2012, 19, 209–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, G.; Zhang, M.; Wang, J.; Zhang, K.; Wu, S.; Zhao, X. Epigenetic regulation of myelination in health and disease. Eur. J. Neurosci. 2019, 49, 1371–1387. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Wang, Y.; Zou, S. A Glance at the Molecules That Regulate Oligodendrocyte Myelination. Curr. Issues Mol. Biol. 2022, 44, 2194–2216. [Google Scholar] [CrossRef]
- Wang, H.; Matise, M.P. Tcf7l2/Tcf4 transcriptional repressor function requires HDAC activity in the developing vertebrate CNS. PLoS ONE 2016, 11, e0163267. [Google Scholar] [CrossRef] [Green Version]
- Zeng, C.W.; Kamei, Y.; Wang, C.T.; Tsai, H.J. Subtypes of hypoxia-responsive cells differentiate into neurons in spinal cord of zebrafish embryos after hypoxic stress. Biol. Cell 2016, 108, 357–377. [Google Scholar] [CrossRef]
- Zeng, C.W.; Sheu, J.C.; Tsai, H.J. Hypoxia-responsive subtype cells differentiate into neurons in the brain of zebrafish embryos exposed to hypoxic stress. Cell Transplant. 2022, 31, 09636897221077930. [Google Scholar] [CrossRef]
- Lee, H.C.; Lai, W.L.; Lin, C.Y.; Zeng, C.W.; Sheu, J.C.; Chou, T.B.; Tsai, H.J. Anp32a Promotes Neuronal Regeneration after Spinal Cord Injury of Zebrafish Embryos. Int. J. Mol. Sci. 2022, 23, 15921. [Google Scholar] [CrossRef]
- Baumgart, E.V.; Barbosa, J.S.; Bally-Cuif, L.; Götz, M.; Ninkovic, J. Stab wound injury of the zebrafish telencephalon: A model for comparative analysis of reactive gliosis. Glia 2012, 60, 343–357. [Google Scholar] [CrossRef]
- März, M.; Schmidt, R.; Rastegar, S.; Strähle, U. Regenerative response following stab injury in the adult zebrafish telencephalon. Dev. Dyn. 2011, 240, 2221–2231. [Google Scholar] [CrossRef] [PubMed]
- Skaggs, K.; Goldman, D.; Parent, J. Excitotoxic brain injury in adult zebrafish stimulates neurogenesis and long-distance neuronal integration. Glia 2014, 62, 2061–2079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kroehne, V.; Freudenreich, D.; Hans, S.; Kaslin, J.; Brand, M. Regeneration of the adult zebrafish brain from neurogenic radial glia-type progenitors. Development 2011, 138, 4831–4841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rowitch, D.; Lu, Q.; Kessaris, N.; Richardson, W. An ‘oligarchy’ rules neural development. Trends Neurosci. 2002, 25, 417–422. [Google Scholar] [CrossRef] [PubMed]
- Bertrand, N.; Castro, D.S.; Guillemot, F. Proneural genes and the specification of neural cell types. Nat. Rev. Neurosci. 2002, 3, 517–530. [Google Scholar] [CrossRef] [PubMed]
- Guillemot, F. Cell fate specification in the mammalian telencephalon. Prog. Neurobiol. 2007, 83, 37–52. [Google Scholar] [CrossRef]
- Stolt, C.C.; Lommes, P.; Sock, E.; Chaboissier, M.C.; Schedl, A.; Wegner, M. The Sox9 transcription factor determines glial fate choice in the developing spinal cord. Genes Dev. 2003, 17, 1677–1689. [Google Scholar] [CrossRef] [Green Version]
- Lange, C.; Rost, F.; Machate, A.; Reinhardt, S.; Lesche, M.; Weber, A.; Kuscha, V.; Dahl, A.; Rulands, S.; Brand, M. Single cell sequencing of radial glia progeny reveals diversity of newborn neurons in the adult zebrafish brain. Development 2020, 147, 1855951. [Google Scholar] [CrossRef] [Green Version]
- Gräff, J.; Tsai, L.H. Histone acetylation: Molecular mnemonics on the chromatin. Nat. Rev. Neurosci. 2013, 14, 97–111. [Google Scholar] [CrossRef]
- Montgomery, R.L.; Davis, C.A.; Potthoff, M.J.; Haberland, M.; Fielitz, J.; Qi, X.; Hill, J.A.; Richardson, J.A.; Olson, E.N. Histone deacetylases 1 and 2 redundantly regulate cardiac morphogenesis, growth, and contractility. Genes Dev. 2007, 21, 1790–1802. [Google Scholar] [CrossRef] [Green Version]
- Dovey, O.M.; Foster, C.T.; Cowley, S.M. Histone deacetylase 1 (HDAC1), but not HDAC2, controls embryonic stem cell differentiation. Proc. Natl. Acad. Sci. USA 2010, 107, 8242–8247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tao, Y.; Ruan, H.; Guo, X.; Li, L.; Shen, W. HDAC1 regulates the proliferation of radial glial cells in the developing Xenopus tectum. PLoS ONE 2015, 10, e0120118. [Google Scholar] [CrossRef] [PubMed]
- Arnett, H.A.; Mason, J.; Marino, M.; Suzuki, K.; Matsushima, G.K.; Ting, J.P. TNF alpha promotes proliferation of oligodendrocyte progenitors and remyelination. Nat. Neurosci. 2001, 4, 1116–1122. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the author. 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Zeng, C.-W. Macrophage–Neuroglia Interactions in Promoting Neuronal Regeneration in Zebrafish. Int. J. Mol. Sci. 2023, 24, 6483. https://doi.org/10.3390/ijms24076483
Zeng C-W. Macrophage–Neuroglia Interactions in Promoting Neuronal Regeneration in Zebrafish. International Journal of Molecular Sciences. 2023; 24(7):6483. https://doi.org/10.3390/ijms24076483
Chicago/Turabian StyleZeng, Chih-Wei. 2023. "Macrophage–Neuroglia Interactions in Promoting Neuronal Regeneration in Zebrafish" International Journal of Molecular Sciences 24, no. 7: 6483. https://doi.org/10.3390/ijms24076483